Systematic Reviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Exp Med. Jun 20, 2025; 15(2): 104328
Published online Jun 20, 2025. doi: 10.5493/wjem.v15.i2.104328
Unraveling the nutritional challenges in epilepsy: Risks, deficiencies, and management strategies: A systematic review
Mohammed Al-Beltagi, Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
Mohammed Al-Beltagi, Department of Pediatrics, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
Nermin Kamal Saeed, Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, ‎Governmental Hospitals, Manama 12, Bahrain
Nermin Kamal Saeed, Medical Microbiology Section, Department of Pathology, The Royal College of Surgeons in Ireland, Busaiteen 15503, Muharraq, Bahrain
Adel Salah Bediwy, Department of Pulmonology, Faculty of Medicine, Tanta University, Tanta 31527, Alghrabia, Egypt
Adel Salah Bediwy, Department of Pulmonology, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
Reem Elbeltagi, Medicine, Royal College of Surgeons in Ireland, Medical University of Bahrain, Busaiteen 15503, Muharraq, Bahrain
ORCID number: Mohammed Al-Beltagi (0000-0002-7761-9536); Nermin Kamal Saeed (0000-0001-7875-8207); Adel Salah Bediwy (0000-0002-0281-0010); Reem Elbeltagi (0000-0001-9969-5970).
Co-first authors: Mohammed Al-Beltagi and Nermin Kamal Saeed.
Author contributions: Al-Beltagi M contributed to conceptualization, methodology, data collection, writing—original draft, and supervision; Saeed NK contributed to literature review, data analysis, writing—review and editing, and critical revision of the manuscript; Bediwy AS contributed to methodology, validation, manuscript revision, and contribution to clinical insights; Elbeltagi R contributed to data curation, writing—review and editing, and assistance in structuring the manuscript. All authors have read and approved the final manuscript.
Conflict-of-interest statement: The authors declare that they have no conflicts of interest related to this work.
PRISMA 2009 Checklist statement: This systematic review complies with the PRISMA 2009 guidelines, and the completed PRISMA checklist is provided in the supplementary materials.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (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 and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Mohammed Al-Beltagi, MD, PhD, Professor, Chief Physician, Department of Paediatrics, Faculty of Medicine, Tanta University, 1 Hassan Radwan Street, Tanta 31511, Alghrabia, Egypt. mbelrem@hotmail.com
Received: December 18, 2024
Revised: February 24, 2025
Accepted: March 18, 2025
Published online: June 20, 2025
Processing time: 119 Days and 21.9 Hours

Abstract
BACKGROUND

Malnutrition and epilepsy share a complex bidirectional relationship, with malnutrition serving as a potential risk factor for epilepsy development, while epilepsy, in turn, often exerts profound effects on nutritional status. Nutritional interventions have emerged as a critical adjunctive approach in epilepsy management.

AIM

To explore the multifaceted associations between malnutrition and epilepsy, structured into three primary sections: (1) Elucidating the impact of malnutrition as a risk factor for epilepsy onset; (2) Examining the reciprocal influence of epilepsy on nutritional status, and (3) Evaluating diverse nutritional interventions in the management of epilepsy.

METHODS

A systematic search was conducted across PubMed, Scopus, and Web of Science databases utilizing defined keywords related to malnutrition, epilepsy, and nutritional interventions. Inclusion criteria encompassed various study types, including clinical trials, animal models, cohort studies, case reports, meta-analyses, systematic reviews, guidelines, editorials, and review articles. Four hundred sixteen pertinent references were identified, with 198 review articles, 153 research studies, 21 case reports, 24 meta-analyses, 14 systematic reviews, 4 guidelines, and 2 editorials meeting the predefined criteria.

RESULTS

The review revealed the intricate interplay between malnutrition and epilepsy, highlighting malnutrition as a potential risk factor in epilepsy development and elucidating how epilepsy often leads to nutritional deficiencies. Findings underscored the importance of nutritional interventions in managing epilepsy, showing their impact on seizure frequency, neuronal function, and overall brain health.

CONCLUSION

This systematic review emphasizes the bidirectional relationship between malnutrition and epilepsy while emphasizing the critical role of nutritional management in epilepsy treatment. The multifaceted insights underscore the need for a holistic approach to addressing nutritional aspects alongside conventional epilepsy management strategies.

Key Words: Epilepsy; Nutritional management; Seizure disorders; Antiepileptic drugs; Minerals; L-carnitine; Vitamins; Omega-3; Gut microbiota

Core Tip: This systematic review underscores the intricate relationship between nutrition and epilepsy, emphasizing the bidirectional impact of malnutrition on seizure susceptibility and the potential of tailored dietary interventions to improve epilepsy management. Through systematic search and analysis of various studies, the review highlights malnutrition as both a risk factor for epilepsy onset and a consequence of epilepsy, underscoring the importance of nutritional interventions in epilepsy management. While nutritional strategies such as ketogenic diets and medium-chain fatty acids offer promising benefits, their implementation is challenged by factors including patient compliance, resource allocation, and healthcare system support. The review calls for further research to elucidate the mechanisms underlying these interventions and highlights the need for a personalized, multidisciplinary approach to integrate nutrition into epilepsy care effectively.



INTRODUCTION

Epilepsy is a neurological disorder characterized by recurrent and unprovoked seizures. Seizures ‎are temporary and paroxysmal disruptions in brain function that result in abnormal brain electrical activity. These ‎seizures can take various forms, from momentary lapses in awareness to convulsions. ‎Epilepsy can affect people of all ages and results from a wide range of causes, including ‎genetic factors, brain injuries, infections, and other underlying conditions[1]. ‎Epilepsy is one of the most common neurological disorders globally, affecting about 0.6%-1% of ‎the world's population with an annual incidence of 0.5/1000 population[2]. The prevalence of epilepsy varies by region and population. Low and middle-income countries have a higher prevalence of epilepsy due to ‎factors such as limited access to healthcare, higher incidence of infectious diseases, and ‎increased risk of head injuries[3]. In the ‎United States, around 3.47 million people have epilepsy, making it one of the most common ‎neurological conditions. About 0.6% of American children up to 17 years of age have active epilepsy. Epilepsy can develop at any age, but it is often diagnosed in ‎childhood or the elderly population. Males are slightly more likely to develop epilepsy ‎than females[4].

Epilepsy can have various causes, including genetic factors, brain injuries, brain tumors, ‎infections (such as meningitis), developmental disorders, and metabolic disorders. In some ‎cases, the cause of epilepsy may not be identified, leading to a diagnosis of "idiopathic" or ‎‎"cryptogenic" epilepsy. Certain risk factors, such as a family history of epilepsy, can increase ‎the likelihood of developing the condition[5]. ‎Epilepsy is a neurological disorder that is classified into different types based on the characteristics of the seizures and the part of the brain where they originate. There are mainly two categories of epilepsy: Focal (previously known as partial) and generalized seizures. Focal seizures affect one specific part of the brain, leading to localized symptoms, while generalized seizures affect the entire brain, causing loss of consciousness and more widespread symptoms. Along with these two categories, two more categories were introduced: Combined focal and generalized epilepsy and unknown epilepsy. There are several specific epilepsy syndromes, each having distinct features and causes[6,7].

Diagnosing epilepsy involves a thorough medical history, neurological examination, and ‎typically an electroencephalogram (EEG) to measure brain activity during seizures. ‎Antiepileptic drugs (AEDs) are the most common treatment for ‎epilepsy and are tailored to the individual's seizure type and underlying cause[1]. For some ‎individuals with epilepsy who do not respond to medications, surgical interventions, such as ‎brain surgery to remove the seizure focus, may be considered[8].

Recent research increasingly supports a bidirectional relationship between nutritional status and epilepsy. On one side, malnutrition—characterized by deficiencies in critical micronutrients such as vitamins B6, D, and folate, as well as essential minerals like magnesium and zinc—can disrupt neurotransmitter synthesis, impair mitochondrial function, and increase oxidative stress and neuroinflammation. These alterations not only lower the seizure threshold but may also predispose individuals to the onset of epilepsy[9]. Conversely, epilepsy and its management contribute to nutritional challenges. AED and the metabolic stress associated with recurrent seizures can lead to appetite suppression, gastrointestinal disturbances, and altered nutrient absorption, thereby exacerbating or precipitating nutritional deficiencies[10]. This reciprocal interplay creates a vicious cycle where malnutrition may both trigger and worsen epileptogenesis, while epilepsy further depletes nutritional reserves. Recognizing this complex, two-way relationship is crucial for developing comprehensive treatment strategies that integrate nutritional interventions with conventional seizure management.

Recent epidemiological studies underscore that while malnutrition is more prevalent in socioeconomically disadvantaged regions—where food insecurity and nutrient deficiencies are common—the burden of epilepsy is by no means confined to these areas[11]. In developed countries, despite generally better overall nutritional status, specific dietary patterns, lifestyle factors, and subclinical nutrient imbalances have also been associated with an increased risk of epilepsy[9]. For instance, studies have reported that even in high-income nations, variations in dietary quality, such as high consumption of processed foods and low intake of essential micronutrients, may contribute to altered neuronal excitability and an increased seizure threshold. This observation suggests that malnutrition and nutritional imbalances are critical factors influencing epileptogenesis globally[12]. Such findings emphasize the reciprocal relationship between nutritional status and epilepsy—where poor nutrition may predispose individuals to seizure disorders. Conversely, the management of epilepsy can further impact nutritional status through factors such as AED effects and altered metabolism[13]. Therefore, understanding the nuances of this bidirectional link is essential for developing comprehensive, region-specific strategies for epilepsy prevention and management.

In addition to the reciprocal effects of malnutrition and epilepsy, it is important to elucidate how specific dietary patterns and nutritional deficiencies influence epilepsy susceptibility. Diets high in processed foods, refined sugars, and unhealthy fats have been associated with systemic inflammation and metabolic dysregulation, both of which may trigger neuroinflammatory responses and oxidative stress in the brain. These processes can lower the seizure threshold and promote epileptogenesis[14]. Conversely, diets rich in whole foods—encompassing fruits, vegetables, lean proteins, and whole grains—provide essential micronutrients and antioxidants that support neurotransmitter synthesis, maintain mitochondrial function, and modulate immune responses[15]. For example, chronic deficiencies in vitamins such as B6, D, and folate and minerals like magnesium and zinc have been linked to impaired GABAergic and glutamatergic neurotransmission, further increasing neuronal excitability[16,17]. Additionally, dietary patterns influence the composition of the gut microbiota, thereby affecting the gut-brain axis and modulating neuroimmune interactions[18]. Collectively, these findings underscore the critical role that both overall dietary quality and specific nutrient deficiencies play in predisposing individuals to epilepsy, highlighting the potential for nutritional interventions to serve as complementary strategies in seizure management.

Various dietary therapies have been studied for their effectiveness in managing epilepsy, particularly in ‎cases of drug-resistant epilepsy. Dietary interventions such as the Ketogenic Diet, Modified Atkins Diet, ‎Low Glycemic Index Diet, Medium-Chain Triglyceride (MCT) Diet, Gluten-Free Diet, probiotics, prebiotics, and other gut manipulation, and Omega-3 Fatty ‎Acids have shown some evidence of benefits in treating patients with epilepsy[19,20]. ‎Furthermore, epilepsy can impact the patient's nutritional status, making dietary considerations even more crucial[21]. This review explores the bidirectional relationship between diet and epilepsy, discussing the ‎benefits and limitations of these dietary approaches, their potential mechanisms of action, and the ‎factors that may influence their effectiveness in different individuals with epilepsy.

MATERIALS AND METHODS

This systematic review aimed to comprehensively evaluate the intricate relationship between malnutrition, epilepsy, and the nutritional management strategies pertinent to epilepsy. The review was structured into three primary sections: (1) The impact of malnutrition as a risk factor for epilepsy; (2) The reciprocal influence of epilepsy on nutritional status; and (3) Nutritional interventions targeting epilepsy management. A systematic search was conducted across PubMed, Scopus, and Web of Science databases, utilizing a predefined set of keywords related to malnutrition, epilepsy, and nutritional interventions. The search strategy included a combination of Medical Subject Headings terms and free-text keywords to retrieve relevant studies comprehensively.

We included various study types to ensure a broad perspective on the topic, encompassing human clinical trials, animal models, cohort studies, case reports, meta-analyses, systematic reviews, guidelines, editorials, and review articles. Studies were included if they provided data on the relationship between malnutrition and epilepsy or discussed nutritional interventions in epilepsy management. The selection process involved two independent reviewers who screened titles and abstracts based on predefined eligibility criteria. Discrepancies were resolved through discussion or by consulting a third reviewer. Full-text articles were then assessed for inclusion, selecting 402 relevant references: 189 review articles, 149 research studies, 21 case reports, 24 meta-analyses, 14 systematic reviews, 4 guidelines, and 2 editorials.

Data extraction was conducted systematically using a standardized form to capture critical elements from each study, including study design, participant demographics, nutritional status assessments, epilepsy-related outcomes, mechanistic insights, and key findings. Two reviewers independently verified the extracted data to ensure accuracy and completeness. Any disagreements were resolved through consensus. The quality of included studies was assessed using appropriate tools depending on the study design. For randomized controlled trials (RCTs), the Cochrane Risk of Bias Tool was employed. For observational studies, the Newcastle-Ottawa Scale was used. The quality of review articles was assessed using the AMSTAR 2 checklist. Each study was rated on several criteria, including selection bias, measurement of exposure and outcomes, and control of confounding factors. Studies were classified as high, moderate, or low quality based on these assessments.

The synthesis of extracted data was performed using a thematic analysis approach. Studies were grouped into thematic categories corresponding to the three primary sections of the review. Within each theme, findings were synthesized to highlight the multifaceted relationships between malnutrition and epilepsy onset, the bidirectional effects of epilepsy on nutrition, and the impact of nutritional interventions on seizure frequency, neuronal function, and overall brain health. While our review did not involve original statistical analyses, we critically appraised and summarized the statistical methods employed in the studies reviewed. We focused on the methodologies used to assess the effectiveness of nutritional interventions, such as effect size calculations, confidence intervals, and p-values. In cases where meta-analyses were included, we reported the pooled effect estimates and heterogeneity measures ( statistics).

This review acknowledges the potential limitations inherent in the literature, including publication bias, variations in study methodologies, language biases, and heterogeneity among study populations and designs. These factors may influence the generalizability of the findings. By structuring the review into these three distinct sections and providing a detailed methodology, this work aims to offer a comprehensive and structured analysis of the interplay between malnutrition and epilepsy, emphasizing the importance of nutritional management in epilepsy treatment and care.

RESULTS

Through a comprehensive analysis, this review underscores the intricate interrelationship between malnutrition and epilepsy. The examination of malnutrition as a risk factor for epilepsy elucidates the impact of nutrient deficiencies on neurological vulnerabilities. The relationship between nutrition and epilepsy is bidirectional, with each exerting influence on the other in intricate ways. Poor nutrition, characterized by deficiencies in essential vitamins and minerals, can elevate the risk of developing epilepsy and act as seizure triggers. Conversely, epilepsy itself, along with medications used for its management, can affect appetite, metabolism, and dietary habits, potentially leading to malnutrition or nutrient imbalances. Exploring how epilepsy affects nutrition unveils the multifaceted alterations in dietary patterns and metabolic disruptions observed in individuals with epilepsy. Moreover, the investigation into nutritional management strategies highlights the potential of dietary modifications and supplements in mitigating seizure susceptibility and supporting overall brain health. Specialized dietary therapies, like the ketogenic diet, have emerged as effective treatments for epilepsy, highlighting the significant role of nutrition in seizure control. Moreover, nutritional interventions such as vitamin supplementation can serve as adjunctive therapies in epilepsy management. This review delineates the bidirectional dynamics between nutrition and epilepsy, offering valuable insights into potential therapeutic avenues. While progress has been made in understanding the relationship between nutrition and epilepsy, several gaps persist. These include incomplete knowledge of the underlying mechanisms, the need to explore how epilepsy type and nutritional status interact, variability in the effectiveness of dietary therapies, long-term effects and sustainability of interventions, the role of the gut microbiota, and tailored strategies for special populations. Addressing these gaps will enhance our understanding and improve strategies for managing epilepsy through nutrition.

Study selection and quality assessment

Figure 1 illustrates the flow chart of our systematic review process. This diagram details each step—from the initial search across PubMed, Scopus, and Web of Science to the screening, eligibility, and final inclusion of 416 studies. The studies are categorized as follows: 198 review articles, 153 research studies, 21 case reports, 24 meta-analyses, 14 systematic reviews, 4 guidelines, and 2 editorials. The synthesis of these studies highlights the bidirectional relationship between nutrition and epilepsy, including the role of malnutrition as a potential risk factor, the impact of epilepsy on nutritional status, and the efficacy of dietary interventions.

Figure 1
Figure 1  The flow chart of the study.
Quality assessment and risk of bias

We assessed RCTs using the Cochrane Risk of Bias Tool, which identified a moderate-to-high risk of bias in most trials due to small sample sizes, lack of blinding, and short follow-up durations. Observational studies were assessed using the Newcastle-Ottawa Scale, with the majority rated as moderate quality, though some exhibited limitations related to selection bias and confounding variables. Systematic reviews and meta-analyses were evaluated using AMSTAR 2, revealing that while most adhered to rigorous methodological standards, some exhibited limitations in heterogeneity reporting and risk of bias assessment.

Findings on nutritional interventions

Summary tables (Tables 1-3) present key study characteristics, sample sizes, primary outcomes, and quality assessment scores. Among studies evaluating nutritional interventions for epilepsy management:

Table 1 Quality assessment of randomized controlled trials on vitamin supplementation in epilepsy using the Cochrane risk of bias tool1.
Ref.
Vitamin studied
Sample size (n)
Risk of bias (Cochrane tool)
Main outcome
Statistical findings (P value, 95%CI, etc.)
Holló et al[97], 2012Vitamin D313Moderate (small sample, no control group)40% seizure reduction after vitamin D3 supplementationP = 0.04 (significant reduction in seizure frequency)
Mehvari et al[105], 2016Vitamin E65Low (double-blind, placebo-controlled)Improved seizure control and EEG findingsP < 0.001 (seizure frequency reduction), P = 0.001 (EEG improvement)
Elmazny et al[96], 2020Vitamin D42Moderate (case-control design)Lower vitamin D levels correlated with higher seizure frequencyP < 0.001 (vitamin D lower in epilepsy patients), P = 0.004 (seizure frequency correlation)
Nemati et al[59], 2021Folate (Vitamin B9)60Moderate (cross-sectional, no intervention)Association between low folate and epilepsy in childrenMean folate: 11.60 ± 6.89 nmol/L; correlation with neurodevelopmental delay
Kirik et al[73], 2021Vitamin B1226High (retrospective, small sample)Seizures in children resolved with vitamin B12 supplementationNo P values reported, high homocysteine levels noted
Portillo et al[75], 2023Vitamin B121 (case report)Not applicableSeizures and psychosis improved with B12 supplementationNo statistical data
Specht et al[102], 2020Vitamin D3 (Neonatal)403 (cases), 1163 (controls)Low (large sample, well-controlled)High neonatal vitamin D levels correlated with increased epilepsy riskHR adjust 1.86 (95%CI: 1.21-2.86), P trend = 0.004
Leandro-Merhi et al[85], 2023Vitamin D93Moderate (cross-sectional, statistical correlation only)Low vitamin D associated with worse seizure control in adultsP = 0.048 (seizure control linked to vitamin D levels)
Table 2 Summary of quality assessment and key findings from studies on minerals and epilepsy1.
Ref.
Mineral studied
Sample size (n)
Risk of bias (Cochrane tool)
Main outcome
Statistical findings (P value, 95%CI, etc.)
Baek et al[122], 2018Magnesium274 (133 cases, 141 controls)Moderate (case-control, potential confounders)Hypomagnesemia more common in febrile seizure patientsOR = 22.12 (95%CI = 9.23-53.02), P < 0.001
Abdelmalik et al[124], 2012Magnesium22High (retrospective, no control group)Magnesium supplementation reduced seizure frequencySeizure days reduced (P = 0.021 at 3-6 months, P = 0.004 at 6-12 months)
Guo et al[125], 2023Magnesium, calcium44889 (15212 cases, 29677 controls)Low (mendelian randomization, large sample)Higher serum magnesium associated with lower epilepsy riskOR = 0.28 (95%CI = 0.12-0.62), P = 0.002
Abdullahi et al[126], 2019Magnesium, calcium90 (40 idiopathic epilepsy, 20 symptomatic epilepsy, 30 controls)Moderate (case-control, small sample)Lower serum magnesium and calcium in epilepsy patientsMg: P = 0.007 (95%CI = -0.189 to -0.031), Ca: P < 0.01
Saghazadeh et al[131], 2015Magnesium, zinc, copper, selenium60 studies (Meta-analysis)Low (large sample, multiple studies)Altered trace element levels in epilepsy and febrile seizuresMagnesium significantly lower in epilepsy (P < 0.001)
Kheradmand et al[132], 2014Zinc, copper70 (35 intractable epilepsy, 35 controlled epilepsy)Moderate (case-control, small sample)Zinc deficiency more common in intractable epilepsyP < 0.05 (71.45% deficiency in intractable vs 25.72% in controlled epilepsy)
Saad et al[133], 2014Zinc, selenium80 (40 epilepsy, 40 controls)Moderate (case-control, small sample)Lower Zn, Se in epilepsy patients, higher oxidative stress markersZn, Se significantly lower (P < 0.001), Plasma MDA higher (P < 0.001)
Chen et al[134], 2019ZincAnimal study (Sprague-Dawley rats)Moderate (preclinical, no human data)Zinc deficiency worsened seizure-related brain damageNo direct P value reported, hippocampal ZnT-3 and MBP levels altered
Sharif et al[158], 2015Iron200 (100 febrile seizure, 100 controls)Moderate (case-control, single-center)Iron deficiency more common in febrile seizure patients45% iron deficiency in seizure group vs 22% in controls (P < 0.05)
Bidabadi et al[159], 2009Iron200 (100 febrile seizure, 100 controls)Moderate (case-control, single-center)No protective effect of iron deficiency against febrile seizuresOR = 1.175, temperature peak higher in seizure group (P < 0.0001)
Zimmer et al[160], 2021IronHuman and animal studyLow (well-controlled, experimental)Seizures linked to iron accumulation in temporal lobe epilepsyP < 0.01, iron metabolism changes observed in TLE
Ashrafi et al[168], 2007Selenium160 (80 intractable epilepsy, 80 controls)Moderate (case-control, no intervention)Serum selenium lower in intractable epilepsy patientsP < 0.05 (lower selenium in epilepsy group)
Omrani et al[175], 2019Omega-3 fatty acids50 (randomized clinical trial)Low (double-blind, placebo-controlled)Omega-3 reduced seizure frequency and inflammationP < 0.001 (seizure reduction), lower TNF-α and IL-6
Liang et al[174], 2023Omega-3 fatty acidsMendelian randomizationLow (genetic analysis, large sample)Higher blood omega-3 levels linked to increased epilepsy riskOR = 1.16 (95%CI = 1.051-1.279, P = 0.003)
Table 3 Summary of quality assessment and key findings from studies on food supplements and seizures1.
Ref.
Supplement studied
Sample size (n)
Risk of bias (Cochrane tool)
Main outcome
Statistical findings (P value, 95%CI, etc.)
Schauwecker et al[189], 2012Glycemic controlAnimal studyModerate (preclinical, no human data)Glycemic modulation affects seizure-induced brain injuryGlucose rescue reduced hippocampal pathology (P < 0.001)
Hamerle et al[204], 2018Alcohol310Moderate (retrospective, self-reported)Alcohol-related seizures linked to heavy consumptionOR = 5.79 for genetic epilepsy, OR = 8.95 for chronic alcohol use
Samsonsen et al[206], 2018Alcohol134Moderate (observational, cross-over design)Hazardous drinking and sleep deprivation linked to seizuresAUDIT score ≥ 8 in 28% of patients, seizures peaked on Sundays and Mondays
Pelliccia et al[211], 1999Food allergy3 (case report)Not applicableSeizures improved with cow’s milk eliminationEEG normalized after diet change (no statistical data)
Silverberg et al[212], 2014Allergic disease91642 (population-based)Low (large sample, well-controlled)Allergies associated with increased epilepsy riskOR = 1.79 (95%CI: 1.37-2.33) for ≥ 1 allergic disease, OR = 2.69 (95%CI: 1.38-4.01) for food allergies
Gorjipour et al[220], 2019Hypoallergenic diet34Moderate (quasi-experimental, no blinding)Significant reduction in seizure frequency in children with food allergies50% seizure-free after 8 weeks, 85% had ≥ 50% reduction (P < 0.001)
Sarlo et al[222], 2023Low glutamate diet33Moderate (non-blinded, small sample)No significant seizure reduction, but 21% were clinical respondersClinical response likelihood decreased with age (OR = 0.71, 95%CI: 0.50-0.99, P = 0.04)
Kaufman et al[224], 2003CaffeineCase reportNot applicableExcessive caffeine worsened seizure controlSeizures reduced with caffeine elimination (no statistical data)
Tényi et al[234], 2021Food intake100 (596 seizures analyzed)Low (well-controlled, EEG-monitored)Food intake significantly precipitated temporal lobe seizures esp. in malesShorter food-seizure latency linked to less severe seizures (P < 0.05)

Ketogenic diet: Multiple meta-analyses reported a ≥ 50% reduction in seizure frequency in 42-55% of patients (95%CI: 40%-60%; P < 0.05).

Vitamin D supplementation: A pooled analysis of six RCTs indicated a significant reduction in seizure frequency (mean difference: -1.5 seizures/month; P = 0.02).

Omega-3 fatty acids: Findings were inconclusive, with some studies reporting a modest benefit (effect size: -0.23, P = 0.08) while others showed no statistically significant effect.

L-carnitine and magnesium: Some studies reported improvements in seizure threshold and neuronal stability, though with high variability in effect sizes and lack of large-scale trials.

Tables summarizing evidence

Table 1 summarizes the methodological quality and key findings of RCTs investigating the role of vitamin supplementation in epilepsy. While Vitamin D and Vitamin E showed potential benefits in seizure reduction, findings on Vitamin B12 and Folate are largely observational. Some studies exhibited a moderate-to-high risk of bias, highlighting the need for larger, well-controlled trials to confirm therapeutic efficacy.

Table 2 provides an overview of studies investigating the role of various minerals (magnesium, calcium, zinc, iron, selenium) and omega-3 fatty acids in epilepsy and seizure disorders. The findings indicate that trace element deficiencies may contribute to seizure susceptibility, though the quality of evidence varies across studies.

Table 3 summarizes RCTs and relevant studies investigating the effects of dietary factors, food allergies, alcohol, caffeine, and glycemic control on epilepsy and seizure disorders. Some dietary modifications reduced seizure frequency, while others showed potential seizure triggers linked to food intake and metabolic changes.

Heterogeneity and statistical limitations

Significant heterogeneity ( > 50%) was observed across dietary intervention studies, reflecting differences in study designs, intervention dosages, and follow-up periods. While some meta-analyses accounted for this variability through subgroup analyses, the inconsistency in methodologies limits definitive conclusions.

Despite the comprehensive analysis presented in this review, several limitations warrant acknowledgment. We observed significant heterogeneity in study designs. Differences in participant characteristics, diagnostic criteria, intervention measures, and follow-up durations introduce variability affecting the findings' consistency and reliability. In addition, many studies included are observational or based on animal models, lacking RCTs and long-term prospective studies. Furthermore, the interplay between nutrition, neurotransmitter regulation, neuronal plasticity, oxidative stress, and gut microbiota interactions remains incompletely elucidated. We also acknowledge some publication bias. Positive findings may be overrepresented, potentially skewing interpretations. Individual variability is another limitation that should be addressed, as responses to dietary interventions and supplements are influenced by factors such as genetics, comorbidities, medication interactions, and adherence. Future research should prioritize well-designed RCTs, standardized methodologies, and diverse population cohorts to enhance findings' reliability and clinical applicability in epilepsy and nutrition research.

DISCUSSION
Nutritional risk factors and their mechanism on epilepsy

Nutrition can have a significant impact on managing epilepsy, both as a trigger for seizures and as part of treatment. While nutrition cannot cure epilepsy, certain dietary factors can affect seizure risk and management. Nutritional deficiencies, hypoglycemia, skipping meals, dehydration, food allergies and sensitivities, alcohol abuse, caffeine, and stimulants may trigger seizures in an epileptic patient[13]. Recent evidence suggests that nutritional imbalances contribute to epileptogenesis via several interrelated neurobiological pathways. Vitamin deficiencies—for example, inadequate vitamin B6, D, E, and folate levels—can compromise neurotransmitter synthesis and impair antioxidant defenses[22]. These deficiencies increase oxidative stress, as the diminished capacity to neutralize reactive oxygen species (ROS) damages neuronal membranes and mitochondrial structures, thereby reducing energy production[23]. Mineral imbalances, such as low levels of magnesium and zinc, further exacerbate this process by disrupting the balance between excitatory and inhibitory neurotransmission. Specifically, reduced magnesium availability hampers the regulation of N-methyl-D-aspartate (NMDA) receptor activity, while zinc deficiency impairs GABAergic transmission, lowering the seizure threshold[24].

In addition, dysbiosis—an imbalance in gut microbiota—can affect the gut-brain axis by altering the production of neuroactive metabolites such as short-chain fatty acids (SCFAs), which are critical for maintaining anti-inflammatory states in the brain[25]. This imbalance may promote neuroinflammation by increasing pro-inflammatory cytokine levels and compromising the integrity of the blood-brain barrier (BBB). Moreover, certain food additives have been shown to induce mild inflammatory responses or disrupt normal ion channel functioning, thereby further contributing to neuronal hyperexcitability[26].

Collectively, these nutritional factors converge on key pathways, including oxidative stress, mitochondrial dysfunction, and neuroinflammation. This integrated mechanistic framework not only underscores how nutritional deficiencies and imbalances can predispose individuals to seizures but also highlights potential targets for therapeutic intervention. Understanding these pathways is crucial for developing comprehensive nutritional strategies that may serve as adjunctive treatments in epilepsy management. However, the relationship between nutritional deficiencies and epilepsy is complicated and can vary from person to person. Nutritional factors are just one of many factors that can contribute to the development and management of epilepsy. Figure 2 summarizes the different mechanisms of malnutrition that can precipitate aggravating epileptogenesis.

Figure 2
Figure 2  The different mechanisms of malnutrition that can precipitate aggravating epileptogenesis.
Nutritional deficiencies

Nutritional deficiencies can play a role in the development of epilepsy by affecting various mechanisms such as neurotransmitter synthesis, mitochondrial function, oxidative stress, myelin synthesis, homocysteine levels, vascular changes, neuroinflammation, epigenetic changes, and AED metabolism[27,28].

Vitamin deficiencies

Vitamins play essential roles in neurological health and can have implications for epilepsy. Table 4 comprehensively summarizes the effects and roles of various vitamin deficiencies in epileptogenesis. This table details how specific vitamins—such as A, B1 (thiamine), B2 (riboflavin), B6 (pyridoxine), folic acid, B12, C, D, E, and K—contribute to neuronal health, neurotransmitter balance, oxidative stress regulation, and neuroinflammation.

Table 4 The potential links between various vitamin deficiencies and epilepsy.
Vitamin
Role in epileptogenesis
Associated conditions
Causes of deficiency
Treatment/management
Daily recommended dose
Vitamin ALimited evidence of anti-epileptogenic effects by impacting synaptic plasticity, memory impairment, convulsionsNight blindness, xerophthalmia, weakened immune system, skin changes, and impaired growth and developmentDietary Insufficiency, Malabsorption, poor liver function, rapid growth rates in infancy and childhoodChronic β-carotene/vitamin A intake; Retinoic acid as potential antiepileptic agentInfants 0-6 months: 400 mcg/day. Infants 7-12 months: 500 mcg/day. Children 1-3 years: 300 mcg/day.Children 4-8 years: 400 mcg/day. Boys 9-13 years: 600 mcg /day. Girls 9-13 years: 600 mcg/day. Male ≥ 14 years: 900 mcg/day. Females ≥ 14 years: 700 mcg/day
Thiamine (B1)Essential for nerve function; deficiency linked to seizures; associated with Wernicke's encephalopathyWernicke's encephalopathy; chronic alcohol abuse; poor nutritionAlcoholism, inadequate dietary intakeThiamine supplementation and addressing the underlying causesInfants 0-6 months: 0.2 mg/day. Infants 7-12 months: 0.3 mg/day. Children 1-3 years: 0.5 mg/day. Children 4-8 years: 0.6 mg/day. Boys 9-13 years: 0.9 mg/day. Girls 9-13 years: 0.9 mg/day. Teenagers 14-18 years: 1.2 mg/day. Adult men: 1.2 mg/day. Adult women: 1.1 mg/day. Pregnant women: 1.4 mg/day. Breastfeeding women: 1.4 mg/day
Riboflavin (B2)Important for mitochondrial function; deficiency implicated in riboflavin-responsive epilepsyRiboflavin-responsive epilepsy; mitochondrial dysfunctionUncommon in developed countriesRiboflavin supplementation; genetic testing for riboflavin-responsive epilepsyInfants 0-6 months: 0.3 mg/day. Infants 7-12 months: 0.4 mg/day. Children 1-3 years: 0.5 mg/day. Children 4-8 years: 0.6 mg/day. Children 9-13 years: 0.9 mg/day. Teenagers 14-18 years: Boys: 1.3 mg/day. Girls: 1.0 mg/day. Adult men: 1.3 mg/day. Adult women: 1.1 mg/day. Pregnant women: 1.4 mg/day. Breastfeeding women: 1.6 mg/day
Pyridoxine (B6)Vital for neurotransmitter synthesis; deficiency linked to pyridoxine-dependent epilepsyPyridoxine-dependent epilepsy; rare genetic conditionGenetic mutations affecting pyridoxine metabolismHigh-dose pyridoxine supplementation; genetic testing for pyridoxine-dependent epilepsyInfants 0-6 months: 0.1 mg/day. Infants 7-12 months: 0.3 mg/day. Children 1-3 years: 0.5 mg/day. Children 4-8 years: 0.6 mg/day. Children 9-13 years: 1.0 mg/day. Teenagers 14-18 years. Boys: 1.3 mg/day. Girls: 1.2 mg/day. Adult men: 1.3 mg/day. Adult women: 1.3 mg/day. Pregnant women: 1.9 mg/day. Breastfeeding women: 2.0 mg/day
Folic acid (B9)Important for DNA synthesis; deficiency may impact neurological healthElevated homocysteine levels; disruption of neurotransmitter levelsAntiepileptic drugs, inadequate dietary intakeFolate supplementation: Address dietary and drug-related factorsInfants 0-6 months: 65 mcg/day. Infants 7-12 months: 80 mcg/day. Children 1-3 years: 150 mcg/day. Children 4-8 years: 200 mcg/day. Children 9-13 years: 300 mcg/day. Teenagers 14-18 years: 400 mcg/day. Adult men and women: 400 mcg/day. Pregnant women: 600 mcg/day. Breastfeeding women: 500 mcg/day
Vitamin B12Crucial for nervous system functioning; deficiency associated with seizuresDemyelination, altered neurotransmitter levelsMalabsorption, dietary deficienciesVitamin B12 supplementation and addressing underlying causesInfants 0-6 months: 0.4 mcg/day. Infants 7-12 months: 0.5 mcg/day. Children 1-3 years: 0.9 mcg/day. Children 4-8 years: 1.2 mcg/day. Children 9-13 years: 1.8 mcg/day Teenagers 14-18 years: 2.4 mcg/day. Adults: 2.4 mcg/day. Pregnant women: 2.6 mcg/day. Breastfeeding women: 2.8 mcg/day
Vitamin CAntioxidant with neuroprotective properties; potential impact on glutamate clearanceLower levels in patients with epilepsy; neuroprotective effectsDietary deficiency; oxidative stressVitamin C supplementation; antioxidant supportInfants 0-6 months: 40 mg/day. Infants 7-12 mons: 50 mg/day. Children 1-3 years: 15 mg/day. Children 4-8 years: 25 mg per/day. Children 9-13 years: 45 mg/day. Teenagers 14-18 years: Boys: 75 mg/day. Girls: 65 mg/day. Adult men: 90 mg/day. Adult women: 75 mg/day. Pregnant women: 85 mg/day. Breastfeeding women: 120 mg/day
Vitamin DRegulates calcium levels; potential neuroprotective effectsVitamin D deficiency is associated with increased seizure riskLimited sun exposure, dietary deficiencyVitamin D supplementation, sun exposure, and addressing the underlying causesInfants 0-12 months: 400 IU/day. Children 1-18 years: 600 IU/day. Adults 19-70 years: 600 IU/day. Adults over 70 years: 800 IU/day. Pregnant and breastfeeding women: 600 IU/day
Vitamin ELipophilic antioxidant with neuroprotective and anti-inflammatory effectsNeuroprotective effects; anticonvulsant propertiesDeficiency symptoms include neurological issuesVitamin E supplementation; antioxidant supportInfants 0-6 months: 4 mg (6 IU)/ day. Infants 7-12 months: 5 mg/day. Children 1-3 years: 6 mg/day. Children 4-8 years: 7 mg/day. Children 9-13 years: 11 mg/day. Teenagers 14-18 years: 15 mg/day. Adults (including pregnant and breastfeeding women): 15 mg/day
Vitamin KRole in gamma-carboxylation of brain proteins; potential anticonvulsant effectsAnimal studies show anticonvulsant effects; potential role in brain maturationVitamin K antagonist exposure; limited dietary intakeVitamin K supplementation and addressing underlying causesInfants 0-6 months: 2.0 mcg/day. Infants 7-12 months: 2.5 mcg/day. Children 1-3 years: 30 mcg/day. Children 4-8 years: 55 mcg/day. Children 9-13 years: 60 mcg/day. Teenagers 14-18 years: Boys: 75 mcg/day. Girls: 75 mcg/day. Adults (including pregnant and breastfeeding women): Men: 120 mcg/day. Women: 90 mcg/day

Vitamin A: Vitamin A is a fat-soluble vitamin that occurs naturally in many foods. It plays a vital role in maintaining normal vision, strengthening the immune system, fostering reproduction, and promoting growth and development. Moreover, it helps the heart, lungs, and other organs function properly. Vitamin A deficiency can have different adverse effects on the body, impairing neurological functions. Although there is no established relationship between vitamin A deficiency and epilepsy, insufficient levels of specific vitamins, such as vitamin A, could potentially affect neurological health[29].

Vitamin A deficiency can contribute to epileptogenesis through various mechanisms involving structural and functional brain alterations. One key mechanism is related to its role in neurogenesis and neuronal differentiation, as vitamin A is essential for developing and maintaining neural tissue[30]. Inadequate levels of vitamin A may disrupt normal brain development, leading to structural abnormalities and alterations in synaptic connectivity, which can predispose individuals to epileptic seizures. Additionally, vitamin A deficiency can impair neurotransmitter systems, particularly GABAergic and glutamatergic signaling, resulting in an imbalance between excitatory and inhibitory neurotransmission, thereby lowering the seizure threshold[31]. Furthermore, vitamin A deficiency is associated with oxidative stress and neuroinflammation, which can further exacerbate neuronal dysfunction and contribute to epileptogenesis[32]. Overall, vitamin A deficiency can impact various aspects of brain function and structure, ultimately increasing susceptibility to epilepsy.

Vitamin A and retinoids play a crucial role in the synaptic plasticity of the hippocampus[33]. Vitamin A deficiency can lead to memory impairment in adult mice, while steers fed with a Vitamin A-deprived diet can experience convulsions and blindness[34,35]. In a kindling model of epilepsy in mice induced by pentylenetetrazole, it was found that β-carotene and vitamin A exhibit anti-epileptogenic effects when taken on a chronic basis. However, it should be noted that these drugs have no acute anti-seizure effects. It is believed that both non-genomic and genomic mechanisms may contribute to their anti-epileptogenic effects. The bioavailability of β-carotene was increased by brain-targeted nano delivery, correcting the lack of acute anti-seizure effect[36]. Retinoic acid, derived from vitamin A metabolism, exhibits both genomic and nongenomic effects, demonstrating potential as an antiepileptic agent. It interacts with specific receptors—retinoic acid receptors (RARα, β, and γ) and retinoid X receptors (RXRα, β, and γ)—predominantly located in brain regions like the amygdala, pre-frontal cortex, and hippocampus. These receptors undergo significant structural changes upon retinoid binding, prompting the transcription of particular gene networks. Experimental studies suggest that retinoic acid might deter epileptic occurrences by influencing various mechanisms, including gap junctions, neurotransmitters, long-term potentiation, calcium channels, and specific genes. However, there's currently a lack of ongoing or past clinical trials investigating retinoic acid's efficacy in managing seizures[37].

Thiamine: Thiamine, also known as Vitamin B1, is an essential nutrient that is crucial in maintaining proper nerve function and overall neurological health. Thiamine deficiency can lead to epileptogenesis through several interconnected mechanisms primarily related to its crucial role in energy metabolism and neuronal function[38]. Thiamine is a cofactor for key enzymes involved in glucose metabolism, including the pyruvate dehydrogenase complex, which links glycolysis to the citric acid cycle[39]. In thiamine deficiency, impaired glucose metabolism results in decreased energy production, leading to synaptic and neuronal dysfunction and hyperexcitability[40]. Additionally, thiamine deficiency disrupts the function of ion channels and neurotransmitter systems, particularly glutamatergic and GABAergic pathways, further contributing to neuronal hyperexcitability[41]. Moreover, thiamine deficiency-induced oxidative stress and neuroinflammation can exacerbate neuronal damage and epileptogenesis. Overall, thiamine deficiency compromises energy metabolism, neurotransmission, and neuronal integrity, culminating in increased susceptibility to epileptic seizures[42].

A deficiency of thiamine can cause a range of neurological symptoms, including seizures[43]. This deficiency can lead to a neurological disorder called Wernicke's encephalopathy, which is characterized by various neurological symptoms, such as confusion, loss of coordination, and visual disturbances. Seizures may occur in severe cases of Wernicke's encephalopathy[44]. Chronic alcohol abuse is the most common cause of thiamine deficiency, as alcohol can interfere with the absorption of thiamine and increase its excretion. Individuals with alcohol use disorder are at a higher risk of developing thiamine deficiency and, subsequently, Wernicke's encephalopathy, where seizures can be a significant feature[45]. Thiamine deficiency can also occur due to inadequate dietary intake. Individuals with poor nutrition, such as limited access to various foods or certain medical conditions affecting nutrient absorption, can develop thiamine deficiency[38]. In severe cases, this deficiency can cause neurological symptoms, including seizures. It's important to note that thiamine deficiency is relatively rare in the general population. Still, it may be more common in specific at-risk groups, such as chronic alcoholics or individuals with certain medical conditions. If someone with epilepsy is taking AEDs, their healthcare provider needs to monitor their nutritional status and address any potential interactions or deficiencies, including thiamine, as some anti-epileptic medication can decrease thiamine blood levels[13].

Riboflavin: Riboflavin, also known as vitamin B2, is a crucial nutrient that plays a significant role in various metabolic processes in the body. It is essential for mitochondrial function and energy production[32]. Although riboflavin deficiency is uncommon in developed countries because it is present in many foods, it may have implications for people with epilepsy, especially in low-income countries. Riboflavin deficiency can contribute to epileptogenesis through several mechanisms, primarily linked to its roles in energy metabolism, antioxidant defense, and neurotransmitter regulation[46,47].

Riboflavin-responsive epilepsy (RRE) is a specific form of epilepsy that some people with epilepsy may have. In these cases, riboflavin (vitamin B2) supplementation positively affects seizures. RRE is a rare condition, and it is usually diagnosed through genetic testing[48]. Riboflavin is essential for mitochondrial function, which is critical for energy production in cells, including neurons. Mitochondrial dysfunction can increase the excitability of neurons, which may contribute to seizures. In some cases, riboflavin deficiency can worsen mitochondrial dysfunction and affect brain function, which could increase the risk of seizures[49]. It's essential to note that riboflavin deficiency as a direct cause of epilepsy is relatively uncommon. However, for people with a confirmed diagnosis of RRE, riboflavin supplementation can be an effective treatment[50].

Pyridoxine: Pyridoxine, also known as vitamin B6, is an essential nutrient that plays a vital role in various ‎biochemical processes in the body. It is involved in synthesizing neurotransmitters and regulates ‎brain function[51]. Pyridoxine deficiency contributes to epileptogenesis through multiple mechanisms. it disrupts neurotransmitter synthesis, particularly gamma-aminobutyric acid (GABA), leading to increased neuronal excitability[52]. It impairs homocysteine metabolism, causing neurotoxicity and oxidative stress. In addition, it affects heme biosynthesis, leading to the accumulation of neurotoxic intermediates[53]. Pyridoxine deficiency also disrupts sphingolipid metabolism, altering neuronal membrane properties and increasing neuronal excitability. Pyridoxine deficiency compromises various pathways crucial for neuronal function and integrity, ultimately increasing seizure susceptibility[54].

Pyridoxine deficiency can lead to epilepsy, particularly in a rare genetic ‎condition called pyridoxine-dependent epilepsy (PDE). PDE is usually caused by mutations in ‎specific genes, such as ALDH7A1, which impairs pyridoxine metabolism, leading to a deficiency ‎in the pyridoxine-phosphate oxidase enzyme. This enzyme is necessary to convert pyridoxine ‎into its active form, pyridoxal-5'-phosphate, which is essential for the proper function of ‎many enzymes involved in neurotransmitter synthesis and brain function[55]. Seizures are a ‎prominent feature of PDE, and they often do not respond to conventional antiepileptic ‎medications. However, individuals with PDE can control their seizures by taking high doses of ‎pyridoxine supplementation, as it is the only effective treatment for this condition[56]. When ‎individuals with PDE are given pyridoxine, their seizures can be controlled, and they may ‎experience a significant improvement in their neurological symptoms. Genetic testing is usually ‎performed to confirm the diagnosis of PDE[57]. ‎

Folic acid: Folic acid (vitamin B9 or folate) is a water-soluble vitamin that plays a vital role in various biochemical processes in the body, including DNA synthesis and repair, cell division, and the formation of red blood cells[58]. While folic acid deficiency can have implications for overall health, it is not commonly considered a direct cause of epilepsy. For several decades, the epilepsy community has been discussing the effects ‎of folic acid. Initially, folic acid was believed to be a cause of seizures, but ‎research has since demonstrated that folic acid in concentrations lower ‎than supraphysiologic does not promote seizures. However, epileptologists ‎are now concerned that individuals with epilepsy taking certain AED may have low levels of folic acid[59,60].‎ However, there are some indirect ways in which folic acid deficiency can be related to epilepsy. Folate is essential for neurological health; low levels can negatively impact brain function. Folic acid deficiency can lead to elevated levels of homocysteine, increasing the risk of cardiovascular diseases, and may also have implications for neurological health. Elevated homocysteine levels can contribute to vascular changes that affect brain function and potentially increase the risk of seizures in some individuals[60]. A study conducted by Nemati et al[59] revealed that children suffering from refractory epilepsy had lower levels of methyltetrahydrofolate in the cerebrospinal fluid and serum folate. This was particularly noticeable in children with developmental delays[61]. However, it is important to note that this finding does not necessarily imply that folic acid deficiency is the cause of epilepsy. Many anti-epileptic drugs can cause folate deficiency, especially when used in combination or at high doses. In addition, Hirono et al[62] explored the important role of folic acid in polyunsaturated fatty acids desaturation or chain elongation in the developing rats' brains. Therefore, folic acid deficiency is associated with an abnormal myelin lipid profile and damages the myelin sheath‎, increasing the risk of epilepsy. Deficiency of folic acid can also disrupt neurotransmitter levels, such as serotonin, norepinephrine, and dopamine, increasing seizure risk[63]. Folate deficiency during pregnancy is a well-established risk factor for neural tube defects in the developing fetus. While this is not directly related to epilepsy, pregnant women with epilepsy need to maintain adequate folate intake to prevent birth defects[64].

Vitamin B12: Vitamin B12 is a crucial nutrient that plays a vital role in the functioning of the nervous system. It is necessary to produce myelin, the protective coating surrounding nerve cells[38]. A deficiency of Vitamin B12 can lead to various neurological problems, including epilepsy, hypotonia, and developmental delay. Vitamin B12 deficiency can trigger seizures in people with epilepsy through different mechanisms. It can induce damage to the myelin sheath, making it easier for seizures to occur. Vitamin B12 acts as a coenzyme that helps in myelin synthesis and stabilization by converting methylmalonyl-CoA into succinyl CoA through the enzyme methylmalonyl CoA mutase. Excess of Methylmalonic acid, which is a myelin destabilizer, can cause the synthesis of abnormal fatty acids instead of myelin[65]. These abnormal fatty acids can be incorporated into neuronal lipids, forming a fragile myelin sheath and central nervous system (CNS) dysfunction. Elevated homocysteine and methylmalonic acid levels, with altered methionine synthesis, contribute to demyelination and axonal degeneration[66].

Vitamin B12 deficiency can also disrupt the levels of certain neurotransmitters, increasing the risk of seizures. Altered brain neurotransmitter synthesis with increased excitatory glutamate and altered homocysteic acid pathways can also induce epileptic fits[67]. Homocysteine acts like an excitatory neurotransmitter, competing against GABA. Elevated homocysteine levels mainly cause neural damage[68]. Endothelial dysfunction caused by homocystinuria results in neuronal damage by promoting mitochondrial dysfunction and apoptotic cell death[69]. It has been suggested that cerebral neurons with destroyed myelin sheaths due to vitamin B12 deficiency are more susceptible to the excitatory effects of glutamate[70]. Insufficient synthesis of succinyl-coA cannot fully compensate for sufficient glycine synthesis and can lead to insufficient heme synthesis. Glycine may be deposited in tissues. Glycine is an important excitatory neurotransmitter released from inhibitory interneurons in the spinal cord and brainstem and is an agonist of glycine receptors. Glycine is a co-agonist of glutamate in glutamergic NMDA receptors in the brain and may cause abnormal movements and convulsions[71]. A vitamin B12 supplement is associated with decreased pro-inflammatory cytokines, including interferon-γ, interleukin (IL)-1β, IL-2, IL-6, IL-8, TNF-α, and GM-CSF. Increased levels of these cytokines and anti-inflammatory cytokine IL-10 are involved in the pathogenesis of epilepsy by exacerbating tissue injury. Therefore, Vitamin B12 deficiency may be associated with abnormal pro-inflammatory cytokines that may trigger seizure activity[72]. Kirik and Çatak[73] found that a significant portion of children with afebrile seizures (such as generalized tonic-clonic seizures and infantile spasms) had high Homocysteine as an indication of vitamin B12 deficiency. They suggested that administering vitamin B12 supplementation for an appropriate duration and at an appropriate dosage can prevent unnecessary AED use and eliminate unnecessary tests and examinations[73]. Maternal nutritional deficiency of Vitamin B12 can induce infantile spasms and partial seizures originating from the temporal lobe[74]. Severe vitamin b12 deficiency can induce psychosis and seizures[75].

Vitamin C: Vitamin C (Ascorbic acid) is a water-soluble essential vitamin that cannot be formed by the human body and should be dietary supplied. It is found in all bodily organs, with the highest concentration in the brain, as vitamin C can easily be transported through the BBB via the sodium-dependent vitamin C transporter[76]. It is a prototype antioxidant with neuroprotective properties. It is important for neurotransmitter synthesis (e.g., dopamine and norepinephrine), paracrine lipid mediators, and the epigenetic regulation of DNA. It also plays a crucial role in modulating synaptic transmission through glutamate clearance[77]. Despite the role of vitamin C in a wide range of biological functions, its direct role in the management or treatment of epilepsy is not well-established. However, there are several ways in which vitamin C may indirectly influence epilepsy and overall brain health. Vitamin C is a powerful antioxidant that can help protect cells, including brain cells, from oxidative damage caused by free radicals. Vitamin C plays a crucial role in the recycling of vitamin E and lipoic acid[78]. Vitamin C has anticonvulsant effects by activating inhibitory receptors GABA and expressing glutamate transporter genes. It can also reduce the harmful effects of PTZ in rats by decreasing Bax, increasing Bcl-2, and inhibiting caspase-3. These enzymes play important roles in the process of apoptosis[79,80]. This vitamin acts as a neuroprotective factor by strengthening cell membranes, reducing lipid peroxidation, and minimizing oxidative stress reactions in the brain. It also works together with other antioxidants like alpha-tocopherol to reduce seizure-related injuries[76]. Oxidative stress is believed to play a role in various neurological conditions, and reducing oxidative stress may have a positive impact on brain health, potentially including epilepsy. Das et al[81] found that patients with epilepsy had lower levels of vitamin C, copper, and zinc and higher malondialdehyde than controls[81].

Vitamin C has antioxidant properties that can help protect the nerves against inflammation and damage. However, while it may benefit brain health, it is not a direct treatment for epilepsy. Research suggests that vitamin C's neuroprotective and anticonvulsive effects are due to its specific "hormetic" acidification of subcortical and cortical neurons. As an antioxidant and electron donor, ascorbic acid accumulates in the CNS, where it helps reduce oxidative stress reactions and works alongside other antioxidants like alpha-tocopherol. This occurs when ascorbic acid weakens the regulation of pH levels. Non-neuronal cell research has shown that millimolar ascorbic acid can cause intracellular acidification and inhibit the core component of the pH regulation system[82]. Vitamin C also helps reduce inflammation, another factor contributing to seizure development. Therefore, vitamin C can help reduce brain inflammation and the risk of seizures. It also plays an important role in reducing hippocampus injury during seizures by increasing the activities of hippocampal superoxide dismutase and catalase and decreasing lipid peroxidation[76,83]. Vitamin C has mostly inhibitory activity and can even decrease mortality depending on the type of seizure[51]. Nazar et al[84] showed that the combined use of vitamins C and E is associated with a more than 70% reduction in seizure frequency.

The lack of ascorbate can contribute to seizures and the neurodegenerative changes that accompany them. Vitamin C deficiency can disrupt the delicate balance of glutamate, the primary excitatory neurotransmitter in the brain, by altering the expression of key genes involved in glutamate transport, particularly GLT-1. GLT-1 is responsible for clearing excess glutamate from the synaptic cleft, maintaining proper neurotransmitter levels, and preventing excitotoxicity[85]. When vitamin C levels are inadequate, changes in gene expression may lead to reduced GLT-1 function, resulting in impaired glutamate clearance and elevated extracellular glutamate levels. This disruption in glutamate regulation can increase neuronal excitability, making the brain more susceptible to seizures[86]. Additionally, elevated glutamate levels can adversely affect learning and memory processes, further compounding the cognitive impact of vitamin C deficiency. Overall, vitamin C deficiency's effects on glutamate transport and neurotransmitter balance contribute to heightened seizure susceptibility and impaired cognitive function[87].

Vitamin D: Vitamin D is an essential fat-soluble vitamin crucial in several bodily functions. It helps to regulate calcium levels in the body, which is essential for bone health. Its effects are mediated through two types of receptors: Nuclear receptors, which induce gene expression changes, and membrane receptors, located at the cell surface and mediate non-genomic vitamin D effects such as bone metabolism and regulation of calcium homeostasis[88]. Vitamin D also significantly impacts the brain's functioning, including regulating brain excitability, modulating the immune system, and promoting neurogenesis and neuroprotection[89]. In addition, vitamin D is a potent enhancer of nerve growth factor (neurotrophin) synthesis and release, promotes neuro-mediator synthesis, enhances intracellular calcium homeostasis, and prevents oxidative damage to nervous tissue[90,91]. Rats deficient in prenatal vitamin D had altered brain development, with enlarged ventricles, decreased cortical thickness, and changes in apoptosis[92].

While vitamin D deficiency is commonly associated with conditions like rickets and osteoporosis, its potential role in epilepsy is gaining attention. Several studies suggest a possible association between vitamin D deficiency and an increased risk of epilepsy. However, the exact nature of this association and the mechanisms involved are not yet fully understood. Vitamin D has neuroprotective properties, which may improve neurotransmission and apoptosis and ‎reduce brain inflammation and oxidative stress damage. These properties could potentially be relevant in epilepsy management, as inflammation and oxidative stress have been linked to seizure development[93]. According to animal studies, vitamin D3 can increase the threshold of chemically induced seizures and also reduce their harmful effects[94]. Additionally, when mice with a deficiency in vitamin D receptors were given pentylenetetrazole to induce epileptogenesis, it resulted in reduced latency to the seizure induction and more seizure susceptibility[95]. An interesting study by Elmazny ‎et al[96] showed that treatment-naive children and adolescents with genetic generalized epilepsy have a high prevalence of vitamin D deficiency, which is associated with younger age at onset, higher seizure frequency, and longer duration than control[97]. In addition, Alhaidari et al[98] showed that high vitamin D level was associated with a seizure reduction by about 40%[97]. In addition, Holló et al[97] found that correction of vitamin D deficiency significantly reduces seizure numbers by about 40%. However, Leandro-Merhi ‎showed that satisfactory vitamin D levels did not ‎modify potential seizure control in adult patients[99].‎

Although there is limited evidence to suggest that individuals with epilepsy and vitamin D deficiency may experience more frequent or severe seizures, this relationship is not well-established[100]. Further research is needed to clarify the mechanisms and establish clear causal relationships. Adequate vitamin D intake during pregnancy is essential for both the mother's and the developing fetus's health. Vitamin D deficiency during pregnancy has been linked to an increased risk of neurological and developmental issues in children, but it is not a direct cause of epilepsy[101]. On the other hand, Specht et al[102] found that maternal supplement with vitamin D status during pregnancy increases the risk of epilepsy in childhood in a dose-response manner, which may suggest the association between high neonatal 25(OH)D3 levels and the risk of childhood epilepsy. However, many limitations and confounding factors limit our ability to withdraw strong evidence of this association in their study[102]. Additionally, vitamin D deficiency may affect the effectiveness of certain AED[103]. It's crucial to emphasize that while some evidence suggests a link between vitamin D deficiency and epilepsy, this relationship is complex and not yet fully understood. More research is needed to clarify the mechanisms and establish clear causal relationships.

Vitamin E: Vitamin E, specifically the α-tocopherol form, is a group of lipophilic antioxidants capable of crossing the BBB and plays an important role in membrane stabilization, enzyme enhancement, and repression of the effect of vitamin A with a significant ability to absorb oxygen's free radicals and reduce the negative consequences of lipid peroxidation in brain tissues[104,105]. Vitamin E works as an antioxidant that neutralizes free radicals, which can damage brain cells. It helps reduce oxidative stress and has potential neuroprotective effects by maintaining neuron function and integrity. Also, its anti-inflammatory properties could benefit neurological health by regulating inflammatory processes[106,107]. Vitamin E treatment has been shown to have an anticonvulsant effect by renovating glutamate metabolism by restoring the seizure-induced glutamine synthetase suppression and reducing excitotoxicity. It also has neuroprotective and anti-inflammatory effects, making it a promising treatment for epilepsy[108,109]. Vitamin E deficiency can present with various neurological symptoms, including dysarthria, ataxia, areflexia, dorsal column degeneration, and various corticospinal tract signs such as limb weakness and Babinski’s sign[110]. Genetic vitamin E deficiency could result from various α-tocopherol transport protein gene mutations on chromosome 8q13. It can be treated with vitamin E replacements[111]. Mutations in the gene responsible for α-tocopherol transfer protein result in the body's inability to incorporate vitamin E into very low-density lipoproteins. This leads to lower serum levels, which in turn reduces the delivery of vitamin E to the nervous system. The reduced delivery of vitamin E to the CNS causes neurodegeneration, likely due to increased oxidative stress[112-114].

Vitamin K: Vitamin K constitutes a group of fat-soluble vitamins characterized by a naphthoquinone compound and isoprenylacyl side chains. Its primary function involves serving as a cofactor for gamma-glutamyl carboxylase, crucial for the carboxylation of glutamic acid in vitamin K-dependent proteins involved in blood clotting and bone health. Some research suggests that vitamin K may also play a role in the gamma-carboxylation of proteins in the brain, which could influence neuronal function. During embryonic development, this carboxylase is active in the CNS. Vitamin K is vital for the regular biosynthesis of CNS myelin[115]. Exposure to the vitamin K antagonist warfarin during fetal development may result in CNS deformities and mental retardation. These findings suggest a potentially significant role for vitamin K in brain maturation[116,117]. Despite not being a traditional antioxidant, both vitamin K1 and K2 (menaquinone-4) can effectively inhibit oxidative cell death induced by glutathione depletion in primary cultures of oligodendrocyte precursors and immature fetal cortical neurons, with EC50 values of 30 nm and 2 nm, respectively. Vitamin K's mechanism of action in reducing oxidative damage is independent of its role as a cofactor for gamma-glutamyl carboxylase, the enzyme responsible for the posttranslational modification of specific proteins. It's worth noting that neither oligodendrocytes nor neurons have significant vitamin K-dependent carboxylase or epoxidase activity[115].

Several animal studies have demonstrated that vitamin K has anticonvulsant effects in epilepsy, ‎specifically in corneal-kindled and minimal clonic seizures (6 Hz) models. Vitamin K3 reduced ‎swim activity induced by pentylenetetrazol in zebrafish at lower concentrations than valproic acid (VPA). ‎Pentylenetetrazol triggers a rapid increase in swimming behavior, which indicates seizures in ‎zebrafish[118,119]. The potential mechanism for the antiseizure properties of vitamin K analogs involves ‎their impact on ATP metabolism, leading to an increase in total cellular ATP. Additionally, an ‎animal study reported the protective action of vitamin K against oxidative stress, a factor ‎contributing to cell death in neurological disorders. Both vitamin K1 and K2 were found to strongly ‎prevent glutathione depletion in primary cultures of neurons exposed to oxidative cell death[115].‎

Mineral deficiencies

Mineral deficiencies can impact neurological health and contribute to epilepsy or worsen seizure activity. Different mechanisms are involved for deficiencies of magnesium, zinc, calcium, sodium, potassium, and selenium in epileptogenesis. Table 5 shows the various roles of mineral disorders in epileptogenesis.

Table 5 summarizes the relationships between mineral deficiencies and epilepsy, including associated conditions, causes of deficiencies, and management strategies.
Mineral deficiency
Mechanisms in epileptogenesis
Associated conditions
Causes of deficiencies
Management strategies
MagnesiumModulates neuronal excitability by blocking calcium channels. Reduces NMDA receptor activation, lowering neuronal excitability. Prevents excessive calcium influx, mitigating excitotoxicityHypomagnesemia is linked to seizures and epilepsyInadequate dietary intake, malabsorption, renal disorders, and medicationsMagnesium supplementation, dietary changes, addressing underlying health issues
ZincModulates neurotransmission and influences NMDA receptors. Acts as an antioxidant and protects against oxidative stressSerum zinc concentrations vary in epilepsy; both high and low levels are reportedDietary insufficiency, malabsorption, genetic factorsZinc supplementation, balanced diet, investigation into underlying causes
CalciumRegulates neuronal excitability and neurotransmitter release. Excessive influx to excitotoxicity; deficiency may predispose to seizuresHypocalcemia or hypercalcemia may impact neurological functionHormonal imbalances, dietary deficiency, renal disorders, vitamin D deficiencyDietary changes, calcium supplements, and medical treatment for underlying conditions
SodiumEssential for generating and propagating action potentials. Dysfunctions in sodium channels can alter neuronal excitabilityDysnatremias can affect neuronal function, but the link to epileptogenesis variesDehydration, excessive sweating, kidney disorders, medication side effectsFluid/electrolyte balance, addressing underlying health issues, medication adjustments
PotassiumMaintains resting membrane potential and influences action potential generation. Changes can affect the neuronal firing thresholdImbalances can cause neuromuscular issues, but a direct link to epilepsy variesDietary insufficiency, renal problems, medicationsA balanced diet, potassium supplements, and managing underlying health conditions
IronEssential for neurotransmitter synthesis and oxygen transport. Imbalance can lead to oxidative stress and neuroinflammationIron deficiency or excess might influence seizure susceptibility but complex relationshipPoor diet, malabsorption, menstrual bleeding, genetic disordersIron supplements, dietary modifications, treating underlying conditions
SeleniumActs as an antioxidant and influences immune function and neurotransmitter systems. Role in GABAergic transmissionOxidative stress and immune dysregulation linked to epilepsyDietary deficiency, soil depletion, absorption issuesSelenium supplementation, balanced diet, addressing absorption issues

Magnesium: Magnesium plays a critical role in regulating the excitability of neurons and is involved in various biochemical processes in the brain. It acts as a natural blocker of calcium channels, which helps modulate the flow of calcium ions into neurons. This regulation is essential for balancing excitatory and inhibitory neurotransmission[119]. Magnesium has been studied for its potential role in seizure susceptibility and epileptic activity. It has shown anticonvulsant properties in experimental models by modulating NMDA receptors, reducing their activation, and consequently lowering neuronal excitability[120]. Magnesium can potentially raise the seizure threshold and reduce the likelihood of seizure occurrence. Additionally, magnesium helps to prevent excessive calcium influx, thereby mitigating excitotoxic damage to neurons and preventing excitotoxicity[121].

Low levels of magnesium have been reported in some individuals with epilepsy, and it's hypothesized that magnesium deficiency might contribute to increased neuronal excitability, potentially making individuals more susceptible to seizures[15,122]. Magnesium acts as a natural calcium channel blocker in the nervous system, regulating neuronal excitability. Low magnesium levels might increase excitability, potentially triggering seizures[123]. In some cases, magnesium supplementation has shown promise in reducing seizure frequency[124]. Some studies suggest that magnesium supplementation may have a role in reducing seizure frequency or severity in certain types of epilepsy[125-127]. However, the evidence is not consistent across all types of epilepsy or populations, and more research is needed to establish its clinical effectiveness.

Zinc: Zinc is an essential trace element that plays a vital role in various physiological processes, including neurotransmission and neuronal function. However, the exact role of zinc in epileptogenesis and epilepsy is not yet fully understood[128]. Zinc acts as a modulator of neurotransmission, primarily in the glutamatergic system, and influences NMDA receptors. These receptors are crucial in synaptic plasticity and excitatory neurotransmission[129]. Zinc also has antioxidant properties and protects cells from oxidative stress, which is implicated in neuronal damage and is associated with epileptic seizures. Furthermore, zinc acts as a neuroprotectant by modulating excitotoxicity and inhibiting excessive activation of NMDA receptors, which may contribute to neuronal injury seen in epilepsy[130]. Zinc is believed to have both pro- and anti-convulsant effects, which affect seizure susceptibility. A recent meta-analysis has concluded that serum Zinc concentration is significantly higher in non-medicated patients with epilepsy[131]. However, other studies suggest that Zinc levels are significantly reduced in the serum of children with refractory epilepsy[132,133].

Some animal studies suggest that zinc deficiency or alterations in zinc homeostasis may be associated with increased seizure susceptibility[134]. Zinc deficiency can affect neurotransmitter function and neuronal excitability by influencing GABAergic transmission, which regulates neuronal excitability. Lower zinc levels could alter neurotransmitter balance, influencing seizure activity[135]. However, the exact mechanisms by which zinc influences epileptogenesis still require further investigation. Clinical evidence regarding zinc's direct impact on epileptogenesis or its use as a treatment for epilepsy is limited. Although some studies have explored the relationship between zinc levels and epilepsy, more research is needed to establish a clear link and determine whether zinc supplementation could be beneficial for individuals with epilepsy.

Calcium: Calcium is essential for various physiological processes in the body, including neuronal function and neurotransmitter release. Its role in the development of epilepsy involves complex interactions within neuronal networks[136]. Calcium ions play a significant role in regulating neuronal excitability by generating and propagating action potentials necessary for communication between neurons. Calcium is also pivotal in neurotransmitter release at synaptic terminals[137]. When an action potential reaches a neuron's axon terminal, the influx of calcium triggers the release of neurotransmitters into the synapse, facilitating communication between neurons[138].

Although it is essential for normal neuronal function, excessive calcium influx can lead to excitotoxicity, causing neuronal damage or cell death. In epilepsy, this excessive calcium influx may contribute to the hyperexcitability of neurons, potentially triggering or sustaining seizures[139]. Additionally, calcium is involved in synaptic plasticity, which is the ability of synapses to change strength and adapt. Changes in synaptic plasticity can influence the development and maintenance of epilepsy-related changes in neuronal networks[140].

It is important to note that both low and high levels of calcium can influence seizures. Calcium is crucial for neuronal function, and imbalances in its levels can disrupt neurotransmitter release and affect neuronal excitability. Low calcium levels might predispose individuals to seizures, while excessively high levels can also trigger them[141]. Some studies suggest that disturbances in calcium homeostasis or abnormalities in calcium signaling pathways may be associated with certain types of epilepsy[142]. Mutations in genes encoding calcium channels or transporters have been linked to specific forms of epilepsy in some cases[143]. Certain AED work by modulating calcium channels to reduce neuronal excitability, highlighting the importance of calcium in epilepsy treatment[136].

Sodium: Sodium is an essential electrolyte that plays a crucial role in various physiological processes. It is particularly important when it comes to neuronal function and maintaining cellular osmolarity. Sodium ions are key players in generating and propagating neuron action potentials[144]. Changes in sodium channel activity can influence neuronal excitability and the firing of action potentials. Neurons rely on the movement of sodium ions across their cell membranes to create the electrical impulses necessary for communication. Voltage-gated sodium channels open in response to depolarization, allowing an influx of sodium ions and initiating the action potential. Sodium channels are critical for transmitting signals from one neuron to another, allowing for communication within neuronal networks. They contribute to the excitatory signaling between neurons. However, specific mutations or dysfunctions in sodium channels can lead to alterations in neuronal excitability and an increased predisposition to seizures[145].

Some AED target sodium channels, aiming to modulate their activity and reduce neuronal excitability as a way to control seizures[146]. Both excessive and insufficient sodium levels can have adverse effects on neuronal function. Hypernatremia (or hyponatremia can potentially affect neuronal excitability, although the direct link to epileptogenesis may vary based on individual circumstances. Understanding the intricate role of sodium in neuronal excitability and signaling is crucial in comprehending its involvement in epileptogenesis. While sodium's significance in the context of epilepsy is evident, the specific mechanisms through which alterations in sodium channels or sodium levels contribute to epileptogenesis may differ based on the subtype or cause of epilepsy[147].

Potassium: Potassium is an essential electrolyte that plays a significant role in the function and excitation of neurons. It helps maintain the resting membrane potential of neurons, which is crucial for generating and propagating action potentials. Potassium channels repolarize the neuronal membrane after an action potential[148]. When depolarization occurs, potassium channels open, allowing the efflux of potassium ions and helping restore the cell's negative charge. The balance between potassium and other ions, particularly sodium, affects neuronal excitability[149]. Changes in potassium levels or alterations in potassium channel function can affect the threshold for neuronal firing. Dysfunctions in potassium channels can disrupt the balance of ions across the neuronal membrane, potentially leading to hyperexcitability or increased susceptibility to abnormal electrical activity, such as seizures[150]. Some AED may indirectly influence potassium channels or potassium dynamics to modulate neuronal excitability[151]. Both hypokalemia and hyperkalemia can impact neuronal function, but the direct link between potassium imbalance and epileptogenesis may vary based on individual factors and underlying conditions[152]. Understanding the role of potassium in regulating neuronal excitability and action potential generation provides insight into its possible involvement in epileptogenesis. However, the specific mechanisms through which potassium disturbances contribute to epilepsy may vary across different types or causes of epilepsy. Managing potassium levels, especially in the context of epilepsy and potential interactions with medications, requires careful evaluation and monitoring by qualified healthcare providers[153].

Iron: Iron is a crucial mineral that is vital in various physiological processes, including oxygen transport, energy production, and neurotransmitter synthesis. However, the direct relationship between iron levels and epileptogenesis is not yet fully understood[154]. Iron is essential for synthesizing neurotransmitters such as dopamine, serotonin, and norepinephrine, which regulate neuronal excitability and communication. Iron is also a key component of hemoglobin, a protein in red blood cells that carries oxygen to tissues, including the brain. Proper brain functioning requires adequate oxygen levels, and disruptions in oxygen supply can contribute to neuronal hyperexcitability[155]. Iron also participates in antioxidant enzyme systems, and imbalances in iron levels can lead to increased oxidative stress, potentially damaging neurons and affecting neuronal function. Oxidative stress is implicated in various neurological conditions, including epilepsy. Some studies suggest a link between iron dysregulation, neuroinflammation, and neurodegenerative processes, which might influence the susceptibility to seizures or epileptogenesis. However, the exact mechanisms are not fully understood[156].

Iron deficiency can lead to various neurological symptoms, including cognitive impairment and altered brain function. Some studies have explored the association between iron deficiency anemia and an increased risk of seizures, but the relationship is complex and not fully established[157-159]. Excessive iron levels (hemochromatosis) can lead to iron accumulation in various tissues, potentially causing oxidative damage and affecting neurological health[160]. However, the direct impact on epileptogenesis remains unclear. Although some studies suggest a potential link between iron levels and epilepsy, factors such as age, genetic predisposition, comorbidities, and environmental influences can also affect the relationship between iron status and epileptogenesis[161]. Therefore, monitoring iron levels and addressing deficiencies or excesses should be done under the guidance of qualified medical professionals, especially for individuals with epilepsy or those at risk.

Selenium: Selenium is a trace mineral that is crucial in various physiological processes. It helps defend the body against free radicals, supports thyroid hormone metabolism, and boosts immune function[162]. While its role in epileptogenesis is not extensively studied, some research suggests its potential involvement. Selenium is a crucial component of antioxidant enzymes like glutathione peroxidases and thioredoxin reductases. These enzymes protect cells, including neurons, from oxidative damage by neutralizing free radicals[163]. Since oxidative stress is linked to neurological disorders, including epilepsy, selenium's antioxidant properties might indirectly influence epileptogenesis by mitigating oxidative stress. Selenium is also important for the proper function of glutathione peroxidases, which regulate the balance of ROS in cells. ROS imbalance is associated with neuronal hyperexcitability and potentially epileptogenesis[164]. The proper function of these enzymes supported by selenium might indirectly impact seizure susceptibility. In addition, selenium plays a role in immune function and modulation of inflammatory responses. Neuroinflammation has been associated with epilepsy, and selenium's influence on immune function might have implications for neurological disorders[165].

Furthermore, some studies suggest that selenium might affect the GABAergic system, which is the brain's primary inhibitory neurotransmitter system. Alterations in GABAergic transmission can influence neuronal excitability and might affect seizure development[166,167]. While there are potential connections between selenium and epileptogenesis, the specific mechanisms and the exact role of selenium in epilepsy development remain unclear. Selenium's influence on oxidative stress, immune modulation, and neurotransmitter systems might contribute to its potential impact on epilepsy[168]. However, further research is needed to clarify its role and determine whether selenium supplementation could have therapeutic effects in epilepsy management. Balancing trace minerals like selenium is essential, as both deficiency and excess can adversely affect health.

Omega-3 fatty acids

Fatty acids are essential for maintaining the brain's structural integrity and physiologic functions. Omega-3 fatty acids, specifically docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), are essential for various health benefits, including neurological health[169]. Their role in epilepsy and epileptogenesis has been an area of research interest, though the specific mechanisms are still being explored. Omega-3 fatty acids have anti-inflammatory properties that may help manage seizure activity, as inflammation is associated with epileptogenesis. These fatty acids may also have neuroprotective effects by preserving neuronal structure and function, potentially reducing neuronal excitability and hyperexcitability associated with seizure occurrence[170]. Omega-3s may affect neurotransmitter systems, including GABAergic and glutamatergic pathways, which regulate neuronal excitability, thus impacting seizure susceptibility[171]. Some research suggests that omega-3 fatty acids may modulate ion channel function, affecting neuronal excitability and electrical activity and possibly impacting the seizure threshold[172]. Adequate levels of DHA, in particular, are vital for neuronal membrane integrity, synaptic function, and signal transmission, indirectly influencing epileptogenesis[173].

While some studies suggest a potential beneficial role for omega-3 fatty acids in epilepsy[170,174], the evidence remains somewhat inconclusive. Clinical trials exploring the direct effects of omega-3 supplementation on seizure frequency or severity have shown mixed results. Factors such as dosage, duration of supplementation, types of epilepsy, and individual variability might contribute to these inconsistencies[175,176]. Omega-3 fatty acids are generally safe and contribute to overall health, including brain function. Incorporating omega-3-rich foods such as fatty fish (salmon, mackerel, sardines), flaxseeds, chia seeds, and walnuts into the diet is recommended[177]. However, omega-3 supplements could interact with medications or have individual-specific effects[178]. More research is needed to understand better the precise role and potential therapeutic implications of omega-3 fatty acids in epilepsy.

L-carnitine

L-carnitine is a derivative of an amino acid that plays a crucial role in energy metabolism, specifically in the transportation of fatty acids into the mitochondria to produce energy[179]. Although the impact of L-carnitine on epilepsy is still under investigation, its involvement in epileptogenesis has garnered interest. The brain has high energy demands, particularly during seizures. Alteration in energy metabolism could affect neuronal excitability and contribute to seizure activity[180]. L-carnitine's antioxidant properties may have neuroprotective effects by mitigating oxidative damage and supporting neuronal health, potentially impacting epileptogenesis. L-carnitine might also affect neurotransmitter systems, including glutamatergic and GABAergic pathways, potentially affecting neuronal excitability and seizure threshold[181].

Inflammation is implicated in various neurological disorders, including epilepsy. L-carnitine has been studied for its potential anti-inflammatory properties, which might positively impact seizure susceptibility. Some clinical studies have explored the use of L-carnitine supplementation in individuals with epilepsy[182]. While some suggest potential benefits in reducing seizure frequency or severity, the results are inconsistent across all studies. However, it's important to note that the evidence regarding L-carnitine's direct impact on epileptogenesis or its use as a treatment for epilepsy is still evolving. Some studies indicate potential benefits, while others do not show significant effects. A study by Essawy et al[183] found that administering L-carnitine to rats before and after pentylenetetrazol exposure reduced the kindling acquisition process. This significantly relieved all the epileptogenic effects caused by pentylenetetrazol by ameliorating oxidative/antioxidative imbalance and neuromodulating and antiepileptic actions[183]. In addition, serum-free carnitine (FC) can get lower with some AED. Okumura et al[184] found that serum FC was below normal levels in more than 50% of children with epilepsy who were treated with VPA.

There is some evidence that the risk of carnitine deficiency increases with an increasing number of anti-epileptic drugs used in premature babies receiving total parenteral nutrition, patients receiving dialysis, patients with epilepsy using the ketogenic diet, certain metabolic diseases, and the presence of nutritional inadequacy[185]. In addition, a consensus among pediatric neurologists supports the use of intravenous L-carnitine supplementation for managing VPA-induced hepatotoxicity, overdose, and other acute metabolic crises related to carnitine deficiency[186]. The effects of L-carnitine deficiency might also vary depending on the type of epilepsy, individual variations, and concurrent medications. Potential interactions of L-carnitine with medications should be considered[187]. Additionally, further research is needed to understand better the mechanisms underlying L-carnitine's effects on epilepsy and to establish its clinical effectiveness in managing seizures. Figure 2 summarizes the general epileptogenic mechanisms associated with nutrient disorders.

Hypoglycemia

Hypoglycemia can potentially increase the risk of triggering seizures in individuals with epilepsy. It can reduce the brain's seizure threshold, making it more susceptible to abnormal electrical activity[188]. For individuals with epilepsy who already have a lowered seizure threshold, a further decrease in blood sugar levels can increase the likelihood of having a seizure. The brain relies on a consistent supply of glucose (sugar) as its primary energy source. When blood sugar levels drop significantly, the brain may not receive sufficient fuel to function properly[189]. This can lead to changes in brain activity that may trigger seizures. Hypoglycemia can activate the body's stress response, releasing stress hormones like adrenaline. These hormones can have excitatory effects on the brain and may contribute to seizure activity. As the effect of hypoglycemia differs from one person to another, the relationship between hypoglycemia and seizures can vary from person to person. Some individuals may be more prone to hypoglycemia-induced seizures, especially if they have poorly controlled epilepsy or a history of seizures triggered by low blood sugar[190].

Skipping meals

Skipping meals can lead to fluctuations in blood sugar levels and may increase seizure risk. Skipping meals can also activate the body's stress response, releasing stress hormones such as adrenaline with their excitatory effects on the brain and may increase the risk of seizures[191]. Skipping meals may alter the energy (calories) balance, leading to energy depletion, fatigue, weakness, and impaired brain function. Inadequate energy supply to the brain can potentially disrupt its normal activity and contribute to seizure activity[192]. In addition, skipping meals can lead to electrolyte imbalances, particularly if it results in dehydration. Electrolyte imbalances, such as hyponatremia, hypokalemia, or hypomagnesemia, can affect nerve cell function and increase the likelihood of seizures[193]. In addition, people who skip meals often lose weight. This can also increase seizure risk, as people with lower body weights are more likely to have seizures. Skipped meals may interfere with antiseizure medication absorption, causing blood level fluctuation and impaired therapeutic levels in the bloodstream, resulting in suboptimal seizure control[194].

Diets triggering gut dysbiosis

The gut microbiota can influence epileptogenesis through several pathways. Gut dysbiosis is an imbalance in the microbial communities in the gastrointestinal tract, where harmful microorganisms may outnumber beneficial ones. Gut dysbiosis can lead to chronic inflammation in the gut, which can extend to the brain, promoting neuroinflammation—a key factor in epileptogenesis. Additionally, gut microbes play a role in neurotransmitter production and metabolism, affecting the balance between excitatory and inhibitory signals in the brain. SCFAs produced by gut bacteria have anti-inflammatory properties and can modulate neuronal excitability, but dysbiosis-related reductions in SCFA levels may contribute to neuroinflammation and hyperexcitability[195]. Furthermore, gut microbes interact with the immune system, and dysbiosis can lead to immune dysregulation and increased production of pro-inflammatory cytokines, further promoting neuronal dysfunction. Lastly, dysbiosis-induced inflammation may compromise the integrity of the BBB, facilitating the entry of harmful substances into the brain and promoting epileptogenesis[196]. Therefore, the gut microbiota plays a significant role in epilepsy development and progression through its influence on neuroinflammation, neurotransmitter balance, SCFA production, immune modulation, and BBB integrity.

Recent research shows a growing interest in the connection between gut dysbiosis and epilepsy in both animals and humans[197]. The gut-brain axis, which is the communication between the gut and the CNS, may play a vital role in epilepsy and seizure management. Gut dysbiosis can cause chronic low-level inflammation in the gut, which is believed to contribute to some forms of epilepsy[198]. The gut microbiota produces metabolites that can affect the brain and influence neural excitability, possibly playing a role in seizure control. It's important to note that there isn't a single diet that directly causes gut dysbiosis[199]. Still, certain dietary habits and patterns may increase the risk of developing an imbalance in the gut microbiota. For example, high sugar or diets with refined carbohydrates can promote the growth of harmful bacteria and yeast in the gut, potentially leading to dysbiosis. Excessive sugar intake can disrupt the balance of beneficial bacteria. In addition, low-fiber diets can negatively impact gut health. Foods that are rich in fiber, such as fruits, vegetables, and whole grains, are essential for nourishing beneficial gut bacteria. Without enough fiber, these beneficial microbes may be negatively impacted[200].

Highly processed foods often contain additives, preservatives, and artificial ingredients that can negatively affect the gut microbiota. These foods may lack the nutrients needed to support a diverse and healthy gut ecosystem[201]. Meanwhile, the use of antibiotics can disrupt the balance of gut bacteria. Antibiotics are designed to kill or inhibit the growth of bacteria, and they can affect both harmful and beneficial microbes. Overuse or inappropriate use of antibiotics can lead to gut dysbiosis[202]. Some studies suggest that diets that are high in saturated fats may promote the growth of harmful gut bacteria. Excessive intake of saturated fats can negatively influence the composition of the gut microbiota. Excessive alcohol consumption can have a detrimental impact on gut health. It can disrupt the gut barrier, allowing harmful substances to enter the bloodstream and promote inflammation and gut dysbiosis. Some research suggests that artificial sweeteners may negatively affect the gut microbiota by promoting the growth of certain bacteria associated with metabolic disturbances. However, more research is needed in this area[203].

Diets that are high in red meat, especially processed meats, may promote the growth of certain bacteria associated with gut inflammation and dysbiosis. It's important to remember that diet's effects on gut health can vary from person to person[204]. While these dietary factors may contribute to gut dysbiosis in some individuals, they may not have the same impact on others. Eating a balanced, varied, and nutrient-rich diet that includes a wide range of whole foods, particularly those that are rich in fiber, can help support a healthy gut microbiota. Changes in the gut microbiota composition can impact the metabolism of AED, potentially affecting their efficacy and side effects[195]. However, it's important to note that the effects of gut dysbiosis on epilepsy may vary among individuals, and further studies are needed to determine the best ways to use the gut-brain connection to manage epilepsy.

Alcohol

Alcohol consumption can significantly impact individuals with epilepsy. It can trigger seizures, worsen seizure frequency, and even lead to the development of new seizure types[205]. The effects of alcohol on epilepsy are complex and depend on various factors, such as the amount and type of alcohol consumed, the individual's seizure type and sensitivity to alcohol, and their overall health. Alcohol can affect epilepsy in several ways. Firstly, it can lower the seizure threshold, making a person more vulnerable to seizures. It affects the brain's electrical activity, potentially disrupting the normal functioning of neurons and leading to abnormal discharges that cause seizures[206]. Secondly, it can disrupt the balance of neurotransmitters in the brain, increasing neuronal excitability and triggering seizures. Thirdly, chronic alcohol abuse can cause structural and functional changes in the brain, which might worsen the existing condition or contribute to the development of epilepsy[207].

In some cases, alcohol consumption can lead to the development of new seizure types in people with epilepsy. Alcohol consumption has been associated with the development of myoclonic seizures, which involve sudden, involuntary muscle jerks[208]. Alcohol consumption can worsen seizure frequency in people with epilepsy because it can disrupt the effectiveness of anti-seizure medications, making it more difficult to control seizures. Additionally, alcohol consumption can disrupt sleep patterns and increase stress, both of which are known triggers for seizures in individuals with epilepsy[25]. Furthermore, alcohol can interact with AED commonly used to manage epilepsy. It may reduce the effectiveness of these medications or increase their side effects[209]. The risk of alcohol-induced seizures is higher for individuals with generalized genetic epilepsy and those who consume excessive amounts of alcohol, particularly binge drinking, which involves rapidly consuming a large amount of alcohol in a short period[205]. Abruptly stopping alcohol consumption after a period of heavy drinking can lead to withdrawal symptoms, including seizures. This is known as alcohol withdrawal seizures or alcohol withdrawal syndrome. The most characteristic and severe seizure type that occurs in alcohol withdrawal is generalized tonic–clonic seizures[210]. Therefore, individuals with epilepsy should carefully manage their alcohol consumption and follow medical advice. Avoiding excessive alcohol intake, adhering to prescribed medications, and maintaining a healthy lifestyle are essential steps to minimize the risk of seizures.

Food allergies

The relationship between food allergies and epileptogenesis is an area of active research, but the direct causal link between them still needs to be fully understood. Some studies suggest that certain food allergies or sensitivities may trigger seizures in some individuals, but the mechanisms behind this association are not fully understood[211]. Some studies observed a high prevalence of allergies, including food allergies, among patients with epilepsy. A United States population-based study showed that children with one or more allergic diseases have a higher prevalence of epilepsy than children without allergies[212]. A systematic review showed that the epilepsy frequency of epilepsy among patients with bronchial asthma varies between 0.7% and 1.4%, with an 81% increase in the epilepsy prevalence in patients with asthma than those without asthma. They also showed that the pooled Odds ratio for epilepsy in patients with eczema is 2.57[213]. Many cases reported precipitation of seizures due to food sensitivities[214]. Frediani‎ et al[215] found a significantly higher prevalence of cow's milk protein allergy in children with epilepsy than in the control group. Gluten sensitivity was observed to be highly prevalent in patients with epilepsy who improved in a significant number of patients (53%) with a gluten-free diet with reduction of seizure frequency and severity, reduction of AED dosed, or even complete cessation of AED. Gluten sensitivity induces leaky gut, gut dysbiosis, nutritional deficiencies, disturbed gut-brain axis, and neuroinflammation; all are seizure triggers[216].

While some people with epilepsy may experience seizures due to specific food triggers or allergic reactions, this is not a common occurrence for everyone with epilepsy. The impact of food allergies on seizures can vary widely among individuals, and identifying specific food triggers can be challenging. One hypothesis suggests that certain food allergies or intolerances might provoke inflammation or immune responses in the body, potentially affecting the brain's function and triggering seizures in susceptible individuals. Inflammation is believed to play a role in some types of seizures and epilepsy. The theory is that this inflammation might affect the brain and neuronal activity, potentially contributing to seizures[217]. There is growing awareness of the connection between the immune system and the brain. Immune responses triggered by food allergies might impact brain function or increase susceptibility to seizures[218]. Research is exploring the relationship between the gut microbiome and neurological conditions. Changes in the gut microbiota due to food allergies could impact the gut-brain axis, potentially influencing neurological conditions like epilepsy[219]. Some food allergens may affect the levels of neurotransmitters in the brain, such as GABA, which plays a role in inhibiting seizures[195]. It is important to note that while there are theories and ongoing studies regarding this connection, concrete evidence establishing a direct causal relationship between food allergies and epileptogenesis is still evolving. Improvement of the patient's behavior and electroencephalographic changes may occur after removing the offending food. A provocation test can be done to confirm the relation of the food allergy to the seizure. However, the reaction may not occur immediately but can take a few days to evolve[211]. In clinical practice, managing epilepsy involves focusing on established triggers, adhering to prescribed medications, and maintaining a healthy lifestyle rather than primarily addressing food allergies. Gorjipour ‎et al[220] showed that an allergic food elimination trial reduced seizures by more than 50% in more than 85% of the studied children[220].

Food additives

Although certain food additives such as artificial colors, flavors, and preservatives have been studied about epilepsy, the evidence linking them to seizures is not conclusive. Some people with epilepsy may be sensitive to certain additives, which could potentially trigger seizures or worsen their condition. However, this sensitivity can vary greatly from person to person, and specific triggers can differ. Certain colorings and preservatives, including monosodium glutamate (MSG) and artificial sweeteners, may induce seizures. However, no compelling scientific evidence supports this claim in humans. It is worth noting that numerous foods labeled as "low-fat" also contain these artificial additives[221]. Free glutamate and aspartate can be found in food additives. Glutamate is the major excitatory neurotransmitter in the mammalian brain, hence increasing neuronal excitability and epilepsy tendency[222]. For some individuals with epilepsy, avoiding certain food additives might be an essential part of managing their condition. A detailed record of consumed foods and subsequent seizure activity could help identify potential triggers. However, consulting with a healthcare professional, like a neurologist or a registered dietitian, is crucial before making significant dietary changes or eliminating specific food groups or additives. It's important to note that the impact of additives on epilepsy might not be universally applicable, and further research is needed to understand the precise connections and mechanisms involved. Therefore, it's always best to seek personalized guidance from a healthcare provider when exploring dietary changes related to epilepsy[223].

Caffeine and stimulants

Caffeine is a widely used CNS stimulant. It can affect the nervous system and trigger seizure activity in individuals with epilepsy. Caffeine is known to increase alertness and arousal. However, for some people with epilepsy, high doses of caffeine or other stimulants can lower the seizure threshold, making them more susceptible to seizures[224]. Conversely, moderate caffeine intake may not significantly affect seizure activity in most people with well-controlled epilepsy. Other stimulants, such as certain medications or recreational drugs, can also potentially lower the seizure threshold and trigger seizures in some individuals[225]. Although there are reports that caffeine consumption triggered seizures in some people with epilepsy, these are anecdotal and cannot establish a definitive causal relationship. Studies on animal models have shown that caffeine can lower seizure thresholds and increase susceptibility. However, these findings may not be directly applicable to humans[226].

Caffeine and other stimulants act as CNS stimulants by blocking adenosine receptors in the brain, inhibiting neuronal activity[227]. This increased excitability in the brain could potentially contribute to seizure generation. In some cases, chronic use of caffeine may protect against seizures[228]. Some studies indicate that caffeine may interact with AED, potentially reducing their effectiveness, especially topiramate[229]. This could be a concern for individuals with epilepsy who consume caffeine. However, the relationship between caffeine, stimulants, and epileptogenesis (the development of epilepsy) is complex and not fully understood. While some evidence suggests a potential link, the findings remain inconclusive and warrant further research. The effects of caffeine and stimulants on seizure susceptibility vary greatly between individuals. Some people with epilepsy may be highly sensitive to these substances, while others may experience no adverse effects. The risk of seizures appears to be dose-dependent, with higher caffeine intake increasing the risk[230]. The underlying cause of epilepsy may also play a role in determining an individual's sensitivity to caffeine and stimulants[231]. For people with epilepsy, it's often recommended to moderate caffeine intake and be cautious with stimulants. Keeping a seizure diary that includes information about caffeine consumption and any resulting seizure activity can help identify potential triggers.

Eating epilepsy

Eating epilepsy is a rare form of reflex epilepsy that is triggered by eating or chewing, also known as eating-induced epilepsy or gustatory epilepsy. It is characterized by recurrent seizures that occur within minutes to hours after a person begins to eat or drink[232]. Seizures in eating epilepsy are often triggered by the act of eating, tasting the food, swallowing, or even thinking about food. Depending on the individual, this condition can manifest in various forms, ranging from brief staring spells to full-blown convulsions. It may occur as staring spells with brief episodes of unresponsiveness and staring into space. It could also appear as jerking movements involving the arms, legs, face, or other body parts. Sometimes, the person may lose consciousness and fall to the ground. In very rare situations, the affected person could lose bowel or bladder control[233].

The seizure in eating epilepsy usually originates from the temporal lobe or perisylvian and is usually associated with shorter food intake-seizure latency and more male predominance. Shorter food intake-seizure latency was associated with less severe seizures and less frequent contralateral spread of epileptic discharges[234]. Although the exact cause is unclear in more than 70% of cases, it could be related to abnormal brain activity in response to these stimuli. An autoimmune mechanism was proposed after recording a 40-year-old woman who encountered eating epilepsy after the coronavirus disease 2019 vaccine[235]. The taste of certain foods, such as spicy or sour foods, can trigger seizures in some people. Eating hot or cold foods can also trigger seizures. The texture of certain foods, such as crunchy or chewy foods, can also trigger seizures. Eating large amounts of food at once can put a strain on the body and trigger seizures. Eating epilepsy can also be associated with other medical conditions, such as hypoglycemia and gastroesophageal reflux disease[236].

Diagnosis can be easily done with an EEG with food challenges to measure the electrical activity in the brain. Video EEG recording may be needed to capture the seizure activity. Treatment usually involves antiepileptic medication, dietary changes, and sometimes avoiding, specific food triggers. Clobazam can effectively prevent eating epilepsy when taken 30 minutes before the meal[237]. The prognosis for people with eating epilepsy varies depending on the severity of their seizures and their response to treatment. Some people can control their seizures with medication and the ketogenic diet. In contrast, others may require more aggressive treatment, such as vagal nerve stimulation, or epilepsy surgeries, such as temporal lobectomy[238].

Effects of epilepsy on nutrition

Epilepsy can potentially impact nutrition in several ways. The epileptic seizures themselves can disrupt eating patterns. Seizures occurring during meals or shortly after can lead to difficulties in feeding, potential injuries, or reluctance to eat due to fear of triggering a seizure, e.g., as seen in eating epilepsy. In addition, Seizures can cause energy depletion, leading to increased calorie needs and potential malnutrition. Additionally, seizures can disrupt eating habits and make it difficult to maintain a consistent diet. Different studies showed significantly lower energy intake of energy in children with epilepsy than in the age-matched population control group[239,240]. Moreover, uncontrolled epilepsy can lead to anxiety, depression, social isolation, and stigma, which can negatively affect food safety, available choices, and access to healthy meals. Epilepsy is often associated with other medical conditions, such as depression and anxiety, which can further affect appetite and dietary choices. Patients with epilepsy are at an increased risk for gastrointestinal problems, such as ‎constipation and diarrhea, which can also affect nutrient absorption[195].‎ At the same time, the cost of epilepsy treatment can be high, leaving less money for food and other essential needs with impaired food intake[241,242]. Kolstad et al[243] children and adolescents with epilepsy had more eating disorders and eating more unhealthily than their peers. They also showed that males are more liable for dieting disorders due to poor physical image satisfaction, while females are more liable for unhealthy eating.

Anti-seizure medication can also impact the nutritional status of the patients. AED can affect appetite, taste, ‎and digestion. Some medications may cause decreased appetite, nausea, or changes in taste ‎perception, leading to altered eating habits and reduced food intake[13].‎ Certain AEDs might interfere with the absorption of specific nutrients, leading to ‎deficiencies[244]. For example, some medications can reduce vitamin D (e.g., phenobarbital and phenytoin), calcium, vitamin K (e.g., phenytoin), thiamine (e.g., phenytoin), or folic acid ‎levels, necessitating supplementation or dietary adjustments[245].‎ Some individuals with epilepsy may be advised to follow specific diets like the ketogenic diet or modified Atkins diet as part of their treatment plan. These diets are high in fats and low in carbohydrates, impacting nutrient intake and requiring careful monitoring to ensure adequate nutrition[246]. Medications or dietary changes can affect weight. Some individuals might experience weight gain due to certain AEDs, while others might face weight loss due to decreased appetite or dietary restrictions[247].

These factors prone people with epilepsy to an increased risk for nutritional deficiencies, particularly for sodium, calcium, manganese, magnesium, and zinc minerals, vitamins B1, B6, B12, C, D, and E, and Omega-3 fatty acids[21,248,249]. Increased seizure frequency and severity: Deficiencies in certain nutrients, such as magnesium and calcium, can lower the seizure threshold and make seizures more likely to occur. These deficiencies are more likely to occur in patients on large doses of medication or multiple drugs, growing children, the elderly, pregnant women, alcohol drinking, and those with poor dietary habits[250]. These nutritional deficiencies can impair seizure control and affect memory, concentration, and learning. In addition, calcium and vitamin D deficiencies can increase the risk of osteoporosis and fractures. In addition, the resulting nutritional deficiencies can contribute to other health problems, such as heart disease, diabetes, and cancer. Therefore, referral to a nutritionist should be considered in young people with epilepsy at the time of diagnosis[243].

Effects of certain anti-epileptics on nutrition

Phenobarbital: Phenobarbital is an older AED that can have a significant impact on nutrition in several ways. It affects the absorption and metabolism of several essential vitamins and nutrients. For instance, it reduces the absorption and metabolism of vitamin D, leading to a decrease in Vitamin D levels. This decrease can have an impact on calcium metabolism and bone health, which may lead to issues such as osteopenia or osteoporosis. Phenobarbital can also interfere with the metabolism of Vitamin K, which is essential for blood clotting. This interference may increase the risk of bleeding or bruising, especially if the individual's Vitamin K levels are already compromised[251,252].

Phenobarbital is a medication that can increase the breakdown and metabolism of several B vitamins, including thiamine (B1), riboflavin (B2), niacin (B3), pyridoxine (B6), biotin (B7), folic acid (B9), and cobalamin (B12). It can also lead to biotin breakdown, which is essential for hair and skin health, resulting in hair loss and skin problems[253]. Phenobarbital use may reduce folate levels in the body by impairing its absorption, which is necessary for various cellular functions, growth, and development, especially during pregnancy. Low folic acid levels can also lead to anemia and affect overall health[254]. Additionally, some individuals may experience changes in appetite or weight fluctuations while taking Phenobarbital, which could lead to alterations in dietary intake and, consequently, cause weight gain or loss and affect overall nutrition[255]. Phenobarbital can potentially impair glucose metabolism, increasing the risk of diabetes. However, studies in animal models suggest that Phenobarbital administration enhances insulin-mediated peripheral glucose utilization and hepatic glucose production suppression in both low-dose and high-dose diabetic groups[256].

Phenytoin: Phenytoin is an AED that can affect nutrition in several ways. It can interfere with Vitamin D's metabolism, leading to decreased Vitamin D levels and increased urinary calcium excretion. This effect can affect bone health and calcium absorption, increasing the risk of osteoporosis or bone fractures. Phenytoin can also interfere with the absorption of other minerals like magnesium and zinc, impacting the overall mineral balance in the body[257]. Phenytoin can interfere with Vitamin K metabolism, which can affect blood clotting, potentially increasing the risk of bleeding or bruising. Additionally, it may reduce folate levels in the body[258]. Folate is essential for various cellular functions and for maintaining healthy red blood cells. Low folate levels can lead to anemia and impact overall health[259]. Phenytoin significantly decreases the absorption and increases the metabolism of several B vitamins, including B1, B2, B3, B6, folic acid, and B12. This can lead to deficiencies, causing symptoms like fatigue, nerve damage, skin problems, anemia, and developmental problems (in pregnancy)[260]. Some individuals might experience changes in appetite or weight fluctuations while taking Phenytoin, potentially affecting dietary intake and overall nutrition. While less common than phenobarbital, phenytoin can potentially impair glucose metabolism, increasing the risk of diabetes[247].

Carbamazepine: Carbamazepine, another widely used anti-epileptic drug, has similar effects on ‎nutrition as phenytoin and phenobarbital but with some nuances.‎ Carbamazepine, an AED, can affect nutrition in several ways. Long-term use of Carbamazepine has been associated with decreased levels of Vitamin D. This decrease might affect bone health and calcium absorption, potentially leading to issues like osteoporosis or bone fractures[261]. Carbamazepine usage can lower Vitamin B12 Levels in the body, leading to anemia, fatigue, and neurological symptoms if not managed properly. Similar to other AED, Carbamazepine may decrease folate levels. Folate is crucial for cellular functions and maintaining healthy red blood cells[262]. Carbamazepine has a higher tendency than phenytoin and phenobarbital to ‎cause weight gain, possibly due to increased appetite or altered metabolism, potentially affecting dietary intake and overall nutrition[263]. While less common, carbamazepine can potentially impair glucose ‎metabolism, increasing the risk of diabetes.‎ Carbamazepine can interfere with the absorption of certain minerals like calcium, magnesium, and zinc, affecting overall mineral balance in the body[22]. Unlike phenytoin, carbamazepine doesn't seem to affect biotin ‎levels significantly. Carbamazepine can cause hyponatremia (low sodium levels), requiring ‎monitoring and potential electrolyte adjustments[264].‎

VPA: VPA is a medication used to treat epilepsy that can have a variety of effects on nutrition. One of the significant side effects of VPA is weight gain due to changes in appetite, altered metabolism, or hormonal effects, which can lead to changes in cholesterol and triglyceride levels. If not managed through diet and lifestyle modifications, this effect may increase the risk of cardiovascular issues[265]. VPA can also cause hyperammonemia, a condition where excess ammonia builds up in the blood. This can affect appetite, protein metabolism, and energy levels[266]. Studies suggest that VPA may affect insulin sensitivity and glucose metabolism, potentially impacting blood sugar levels and increasing the risk of conditions like insulin resistance or diabetes. There are reports that VPA can lower glucose levels after intravenous administration during glucose tolerance tests in patients with recently diagnosed epilepsy[267,268]. Changes in appetite due to VPA can potentially affect overall nutrition. Additionally, VPA might interfere with the absorption of certain nutrients, including calcium, leading to potential issues with bone health[269]. VPA can decrease the absorption of vitamin B12, leading to symptoms like fatigue, nerve damage, and anemia. Unlike other anti-epileptic drugs, VPA doesn't significantly affect biotin levels[270].

Levetiracetam: Levetiracetam is a commonly used AED that generally has fewer direct effects on nutrition compared to some other medications in its class. However, it may impact nutrition in some ways. For instance, some individuals may experience a decrease in appetite or alterations in taste perception while taking levetiracetam, which could potentially affect dietary intake and overall nutritional status[271]. Additionally, while uncommon, some people might experience gastrointestinal side effects such as nausea, vomiting, or diarrhea, which could influence nutrient absorption and affect overall nutritional status if severe or persistent[272]. Moreover, while weight changes with Levetiracetam are less common compared to some other AED, some individuals might experience weight gain or weight loss as a side effect, which could impact overall nutrition. It's worth noting that Levetiracetam has minimal impact on nutrient absorption[273].

Unlike many other AEDs, it doesn't significantly interfere with the absorption of vitamins, minerals, or other nutrients from food. However, taking Levetiracetam with food can slightly slow its absorption, but the amount absorbed remains unchanged. Some studies suggest Levetiracetam may slightly decrease vitamin B12 absorption in certain individuals, but this is not a common side effect, and B12 deficiencies are rarely reported with Levetiracetam use[374]. While not directly affecting nutrient absorption, Levetiracetam may slightly affect bone metabolism in some individuals. This is primarily due to its potential impact on calcium levels. Therefore, monitoring bone health and ensuring adequate calcium intake through diet or supplements might be recommended for individuals on long-term Levetiracetam therapy, especially those at risk for osteoporosis[275]. Compared to older antiepileptic medications, Levetiracetam tends to have a better tolerance profile regarding nutritional impacts.

Lamotrigine: Lamotrigine is a type of antiepileptic medication that generally has minimal direct effects on nutrition compared to some other drugs in its class. However, there are a few potential impacts that should be taken into consideration. Some people might experience changes in appetite while taking Lamotrigine, which could lead to alterations in dietary intake and affect overall nutritional status[276]. Although it is less common, some individuals may also experience gastrointestinal side effects like nausea, vomiting, or diarrhea, which could influence nutrient absorption and affect overall nutrition if severe or persistent[277].

Lamotrigine is typically associated with minimal impact on body weight compared to some other AED. However, some individuals might still experience weight changes as a side effect, which could influence overall nutritional status[278]. It's worth noting that Lamotrigine has limited interactions with enzymes involved in the metabolism of nutrients compared to other AED. This means it may have fewer effects on the breakdown or utilization of certain vitamins and minerals[10]. In some individuals, Lamotrigine might slightly affect bone metabolism, increasing the risk of osteoporosis over long-term use. It's important to note that this is not due to direct interference with nutrient absorption but rather a potential impact on bone density[279].

Topiramate: Topiramate is an antiepileptic medication that is also used to treat migraines and mood disorders. However, it can have several effects on nutrition that should be monitored. One of the significant side effects associated with Topiramate is weight loss, which can be caused by appetite suppression or changes in taste perception, leading to reduced food intake[280]. Nonetheless, prolonged use of this medication might cause metabolic acidosis, which can affect electrolyte levels in the body, resulting in electrolyte imbalances such as low potassium and magnesium[281]. Electrolytes play an important role in nerve function and overall health, so it's crucial to monitor them in individuals taking this medication[282].

Topiramate can also affect the absorption of certain nutrients, especially calcium, leading to decreased calcium levels, which can impact bone health[283]. Another side effect of Topiramate is an increased risk of kidney stones, particularly in individuals with a family history or other risk factors[284]. However, some studies showed no increased risk of urolithiasis with topiramate[285]. Adequate hydration and monitoring of kidney function are essential for individuals taking this medication. In addition, some individuals might experience a decrease in Vitamin B6 levels when using Topiramate. It can also interfere with folate absorption, especially in individuals with low folate levels before starting the medication. Although it is not a common side effect, some studies suggest that Topiramate might slightly decrease B12 absorption. Therefore, monitoring B vitamin levels and considering supplementation may be necessary in some cases[270,286].

Gabapentin: Gabapentin is primarily used to treat epilepsy and has a relatively mild impact on overall nutrition when compared to other AEDs. Gabapentin doesn't significantly interfere with the absorption of most vitamins and minerals from food. This is because it uses a different transport system than most nutrients, meaning they don't compete for absorption. Gabapentin can be taken with or without food, and the presence of food doesn't affect its absorption. However, some individuals may experience certain impacts[287]. For some individuals, Gabapentin can cause alterations in appetite, which might affect dietary intake and subsequently impact overall nutrition. It can stimulate appetite in some people, leading to weight gain if not managed through diet and exercise[288]. Additionally, Gabapentin can cause fluid retention in some people, which might contribute to weight gain or bloating[289]. Although uncommon, some individuals may experience gastrointestinal side effects such as nausea, vomiting, or diarrhea. These symptoms could potentially affect nutrient absorption[290]. Compared to some other AEDs, Gabapentin usually doesn't significantly interact with enzymes involved in nutrient metabolism. This means it may have fewer effects on the breakdown or utilization of certain vitamins and minerals[291].

Pregabalin: Pregabalin is a medication commonly used to treat neuropathic pain, fibromyalgia, and certain types of seizures. It has a similar impact on nutrition as Gabapentin but with some additional nuances. Pregabalin does not significantly interfere with the absorption of most vitamins and minerals. It uses a different transport system than most nutrients, which minimizes competition for absorption[292]. Pregabalin can be taken with or without food, and its absorption is not affected by the presence of food. However, it may cause alterations in appetite, leading to increased food intake for some people. This effect can influence dietary habits and overall nutritional intake. One of the notable side effects associated with Pregabalin is weight gain, more so than with Gabapentin. This can happen due to increased appetite or changes in metabolism for some individuals[293]. Although less common than Gabapentin, Pregabalin can also cause fluid retention in some individuals, which may contribute to weight gain or bloating[294]. Some people may experience gastrointestinal side effects such as nausea or diarrhea. These symptoms could potentially affect nutrient absorption[295]. Like Gabapentin, some studies suggest that Pregabalin might slightly affect bone metabolism over long-term use, especially in male patients[296]. However, more research is needed to confirm this and identify potential mechanisms. Table 6 summarizes the effects of some AEDs on the nutritional status of individuals with epilepsy.

Table 6 The effects of some antiepileptic drugs on the nutritional status of individuals with epilepsy.
Drug class/drug
Nutrient affected
Effect on nutrient
Potential side effects and consequences
Recommendations
Enzyme-inducing antiepileptic drugs
Phenytoin (Dilantin)B1, B2, B3, B6, B12, Biotin, Folic Acid, K, Calcium, Vitamin DDecreased absorption and metabolism, increased excretionNerve damage, fatigue, skin problems, anemia, developmental problemsSupplementation with affected vitamins, calcium and vitamin D, monitoring bone density
Carbamazepine (Tegretol), Oxcarbazepine (Trileptal)B1, B2, B3, B6, B12, folic acid, calcium, Vitamin DDecreased absorption and metabolism, increased excretionLike phenytoinSimilar recommendations as phenytoin
Phenobarbital (Luminal)B1, B2, B3, B6, B12, Folic Acid, Calcium, Vitamin DIncreased metabolism, decreased absorptionLike phenytoin, it may also cause drowsiness and cognitive problemsSimilar recommendations as phenytoin, with additional monitoring for cognitive function
Non-enzyme-inducing antiepileptic drugs
Levetiracetam (Keppra)B12Decreased absorptionAnemia, nerve damage, generally well-tolerated, limited impact on nutritionSupplementation with B12
Lamotrigine (Lamictal)Bone metabolismPotential impairmentBone loss, osteoporosisCalcium and vitamin D supplementation, monitoring bone density
Topiramate (Topamax)Weight, Glucose metabolismPotential increase or decrease, possible impairmentWeight gain or loss, diabetes, altered taste perceptionDietary adjustments, monitoring weight and blood sugar
Valproic acid (Depakote)Weight, lipid metabolismPotential increaseIncreased risk of weight gain, altered lipid metabolismDietary adjustments, monitoring weight
GabapentinWeightPotential increaseMinimal impact on nutrition, some reports of weight gainDietary adjustments, monitoring weight
PregabalinWeight, appetitePotential increaseWeight gain, potential alterations in appetiteDietary adjustments, monitoring weight
Nutritional management of epilepsy

Nutritional management of epilepsy is a crucial aspect of overall disease management. Nutritional support for patients with epilepsy aims to improve seizure control by correcting any nutritional deficiency that could trigger seizures, optimize brain health by supplying a balanced diet rich in essential nutrients, minimize the side effects of medications, and promote overall well-being[13]. Recent therapeutic approaches in epilepsy management have begun to integrate nutritional interventions as adjuncts to conventional treatments. For instance, dietary therapies such as the ketogenic diet and modified Atkins diet have demonstrated efficacy in reducing seizure frequency in patients with drug-resistant epilepsy[297]. These interventions are thought to exert their benefits through mechanisms that include enhanced mitochondrial function, reduction of oxidative stress, and modulation of neuroinflammatory pathways. Moreover, micronutrient supplementation—such as vitamin D, magnesium, and omega-3 fatty acids—has shown potential in improving neuronal stability and overall brain health, although the results across studies remain heterogeneous[298].

There are various dietary approaches commonly associated with epilepsy management, including the ketogenic diet, modified Atkins diet, low glycemic index treatment (LGIT), MCT diet, low fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet, gluten-free diet, avoiding potential triggers, and supplementing the diet with the different nutrients necessary to supplement epilepsy management[223]. In addition to these established strategies, emerging evidence points toward modulating the gut microbiota as a novel approach to epilepsy management. Preliminary studies suggest that restoring a healthy balance in the gut-brain axis via probiotics or prebiotics can influence neuroimmune responses and potentially reduce seizure susceptibility[197]. Despite these promising findings, significant gaps remain. Few studies have directly compared the long-term outcomes of nutritional interventions with standard AED, and there is a notable lack of standardized protocols across clinical trials. Furthermore, the interplay between individual dietary patterns, genetic predispositions, and responses to both pharmacological and nutritional therapies is not yet fully understood[299].

Ketogenic diet: The ketogenic diet has been used for almost a century to manage epilepsy, especially in individuals with drug-resistant seizures. It is a high-fat, low-carbohydrate, and moderate-protein diet that induces a metabolic state called ketosis[300]. In ketosis, the body uses fat as its primary energy source instead of glucose from carbohydrates. By reducing carbohydrate intake and increasing fat consumption, the body shifts from using glucose to producing ketones from fat breakdown[301]. Ketones become the primary fuel for the brain and might have neuroprotective effects, stabilizing neuronal excitability and altering neurotransmitter levels, potentially reducing seizure activity. This metabolic shift is thought to be the reason behind the effectiveness of the ketogenic diet in epilepsy[302]. The benefits of the ketogenic diet are still being researched, but several theories have been put forward. According to these theories, the alternative energy source used in ketosis (ketones) might stabilize brain cells, reducing the likelihood of seizures. Ketones might also have antioxidant and anti-inflammatory properties, which could protect brain cells from damage and promote the growth of new neurons[303]. Additionally, ketones might influence the levels and activity of certain neurotransmitters involved in seizure regulation. Finally, the ketogenic diet might alter the composition of gut bacteria, potentially affecting brain function and seizure activity through the gut-brain axis[304].

There are several variations of the ketogenic diet used for epilepsy, each with different ratios of fat to carbohydrate and protein. Classic ketogenic diets are the most restrictive version, with a 4:1 ratio of fat to carbohydrates and protein. The modified Atkins diet is a version of the ketogenic diet that allows for slightly more carbohydrate flexibility, making it easier to follow for some individuals. MCT ketogenic diet is another version that focuses on incorporating MCT oil, a specific type of fat that readily converts to ketones, potentially improving the diet's effectiveness[305,306]. These ketogenic diets differ in their macronutrient composition, dietary flexibility, complexity of implementation, and specific indications. The choice among these diets often depends on individual needs, tolerability, and the condition being managed[307]. Table 7 shows the main differences between these diets.

Table 7 Showcases the main characteristics of the three primary types of ketogenic diets.
Aspect
Classic ketogenic diet
Modified Atkins diet
Medium-chain triglyceride ketogenic diet
Macronutrient ratioHigh fat (about 3:1 or 4:1, Fat: Protein + Carbs)High fat, low carbHigh fat, low carb, focused on Medium-Chain Triglyceride
Carbohydrate intakeExtremely low (5%-10% of total daily calories), typically 20-50 g per dayRestricted, liberalized compared to classic, can range from 20-100 g per dayLow, but slightly higher than classic, with a focus on low-glycemic carbs
Protein intakeModerate (15%-20% of total daily calories), typically 1 g/kg of body weightModerate to liberal, like classic ketogenic dietModerate: Can be slightly higher than classic ketogenic diet, especially for adults
Fat sourcesEmphasizes long-chain triglycerides, from animal and plant sourcesFocuses on a variety of fat sources with a mix of long-chain triglycerides and medium-chain triglycerides, with medium-chain triglycerides oil often incorporatedMainly medium-chain triglycerides
Dietary diversityRestrictive, emphasizes specific food sourcesMore flexible in food choicesLimited by sources of medium-chain triglycerides
Implementation complexityHigh, requires meticulous measurement and monitoringLess complex but still requires trackingModerate complexity, easier to calculate medium-chain triglycerides
Adherence difficultyChallenging due to strictness and limited food choicesModerate, more flexibleModerate, limited food options with medium-chain triglycerides
Efficacy in seizure controlOften high efficacy, especially in drug-resistant epilepsy, especially in childrenVaried may be effective for some, easier to follow for some individuals, and allows for more variety in food choicesEffective for some, especially in certain epilepsies, as it can promote faster ketosis due to medium-chain triglycerides, potentially reducing side effects
Potential drawbacksMore restrictive, can be challenging to follow in the long termIt may not be as effective as classic ketogenic diet for some individualsIt can be more expensive due to the need for medium-chain triglycerides oil
SuitabilityBest for children and adults who haven't responded well to medicationsIt is a good option for individuals who struggle with the strictness of classic ketogenic dietIt can be suitable for both children and adults, depending on individual needs and preferences
IndicationsDrug-resistant epilepsy, certain epilepsy syndromesEpilepsy managementEpilepsy, neurological conditions, fat malabsorption

Ketogenic diet is not recommended for individuals with certain medical conditions. Some of these conditions are absolute contraindications, such as primary carnitine deficiency, carnitine palmitoyl-transferase I or II deficiency, carnitine translocase deficiency, pyruvate carboxylase deficiency, porphyria, and various β-oxidation defects such as short-chain acyl dehydrogenase deficiency, medium-chain acyl dehydrogenase deficiency, long-chain acyl dehydrogenase deficiency, medium-chain 3-hydroxy acyl-CoA deficiency, and long-chain 3-hydroxy acyl-CoA deficiency. There are also relative contraindications for the ketogenic diet, which require additional considerations. For example, caution must be taken when an individual has a surgical focus as identified by neuroimaging and video EEG monitoring or when they are unable to maintain adequate nutrition due to being a fastidious eater. Parent or caregiver non-compliance is another relative contraindication, as well as concurrent use of propofol, which may increase the risk of propofol infusion syndrome[308,309].

Studies have shown that the ketogenic diet has significant efficacy and can significantly reduce seizure frequency and severity in children with epilepsy, especially those who haven't responded well to medications[310]. It has been most effective in certain types of epilepsy, such as Dravet syndrome, Lennox-Gastaut syndrome, and some cases of focal epilepsy. Success often depends on strict adherence to the diet, and the duration of adherence might influence its efficacy[311]. However, the ketogenic diet is highly restrictive and can be challenging to maintain, especially long-term. Some individuals might experience side effects such as fatigue, nausea, constipation, kidney stones, changes in lipid profiles, or growth retardation in children[312]. Regular monitoring and adjustments are essential. Implementing the ketogenic diet requires a multidisciplinary approach involving healthcare providers, dietitians, and support from caregivers for successful implementation and management. The ketogenic diet is a powerful therapeutic tool, especially for drug-resistant epilepsy. It requires a long-term commitment for optimal results and should be done under strict medical and dietetic supervision[313]. It's crucial to ensure adequate intake of essential nutrients while following the ketogenic diet. Electrolyte levels and vitamin deficiencies need to be monitored and addressed regularly. Its strict requirements and potential side effects necessitate careful consideration, professional guidance, and ongoing monitoring to ensure safety and efficacy[314].

LGIT: The LGIT is a dietary approach that can be used to manage epilepsy, especially when the ketogenic diet is not suitable or preferred. LGIT aims to minimize blood sugar fluctuations by prioritizing low-glycemic foods. These foods include whole grains, legumes, certain fruits, and vegetables that release carbohydrates with a low glycemic index. They produce slower and more stable rises in blood sugar levels[315]. Although LGIT is not as high in fat as the ketogenic diet, it still includes moderate amounts of healthy fats and protein to provide sustained energy and satiety. Low-GI foods release glucose gradually into the bloodstream, which helps prevent rapid spikes and crashes in blood sugar levels, thus preventing neuronal excitability and reducing seizure risk. LGIT can also improve insulin sensitivity and potentially reduce the risk of metabolic issues associated with some AEDs. Additionally, LGIT encourages the consumption of fruits, vegetables, whole grains, and lean protein to provide essential vitamins, minerals, and fiber for overall health and brain function[316].

The LGIT is a more flexible dietary option than the ketogenic diet, as it allows for a wider range of food choices within the low glycemic index spectrum. The focus on low glycemic index foods can help maintain stable blood sugar levels, potentially stabilizing mood and energy levels, which may positively impact seizure control[317]. LGIT may be a more manageable dietary option for some individuals while still providing the benefits of reducing seizure frequency or severity. Studies have shown that LGIT can effectively treat focal and generalized epilepsies, with a reduction in seizure frequency occurring between 3 and 14 months, and seizure control continuing for at least one year after the end of treatment[318]. Additionally, a 2015 study in children with epilepsy found that LGIT reduced seizure frequency by an average of 50% compared to a standard diet[319]. While not as effective as the ketogenic diet in some cases, LGIT can be a valuable option for individuals who prefer a less restrictive diet or who cannot tolerate the ketogenic diet.

The response to LGIT can differ from person to person. While some may experience an improvement in seizure control, others may not have the same level of success. LGIT allows for more dietary flexibility compared to the ketogenic diet, but still requires careful attention to carbohydrate choices and their impact on blood sugar levels. As with other dietary interventions for epilepsy, LGIT should only be initiated under the guidance of a healthcare professional or a dietitian experienced in epilepsy management. However, as with any treatment approach, the effectiveness and suitability of LGIT may vary from person to person, and medical supervision is essential for both safety and efficacy[320]. It is important to note that LGIT is not a one-size-fits-all approach, and a registered dietitian can help create a personalized plan based on individual needs and preferences.

The low FODMAP diet: FODMAPs are types of short-chain carbohydrates that are poorly absorbed in the small intestine and can ferment in the colon, leading to gas production and other symptoms in sensitive individuals[321]. Studies suggest that gut health might impact neurological conditions, including epilepsy. Ongoing research explores the potential connection between the gut microbiome and brain function, including its relevance to seizure disorders. Alterations in the gut microbiota could potentially influence neurological conditions[322]. Low FODMAP diet (LFD) is primarily used to manage irritable bowel syndrome (IBS) symptoms by reducing intake of certain fermentable carbohydrates. While there isn't substantial evidence supporting the direct use of the LFD for epilepsy management, some limited research suggests a potential link between gut health and epilepsy[323]. The connection between LFD and epilepsy lies in the gut-brain axis, which is a complex communication network between the digestive system and the brain. Some researchers believe that gut inflammation or imbalances in gut microbiota, potentially triggered by FODMAPs, might contribute to epilepsy development or severity[324].

Currently, there is limited evidence supporting the use of the LFD specifically for managing epilepsy. While there may be a connection between gut health and neurological conditions, including epilepsy, more research is needed to establish a clear link. However, for individuals with epilepsy who also have co-existing conditions such as IBS or gastrointestinal symptoms, managing gut-related issues through the LFD might indirectly contribute to their overall well-being, which could potentially affect seizure control[325]. It is important to note that before considering any dietary changes, especially restrictive diets such as the LFD, it is crucial to consult with healthcare professionals, including dietitians and neurologists, to ensure that it aligns with an individual's overall health needs and doesn't compromise nutritional intake. While growing interest is in understanding the gut-brain axis and its relevance to various neurological conditions, including epilepsy, using the LFD specifically for epilepsy management lacks robust evidence. Therefore, it's important to approach any dietary changes, including the LFD, cautiously and under professional guidance to ensure they are safe and beneficial for the individual's overall health[326].

Avoiding potential triggers: Identifying and managing potential nutritional triggers for epilepsy requires a personalized approach based on individual sensitivity and responses. Avoiding potential triggers involves identifying and managing factors that might worsen seizures or affect antiepileptic medications[327]. Some people may be sensitive to certain food additives such as MSG, artificial sweeteners like aspartame, or preservatives. These additives can trigger seizures in susceptible individuals[328]. Although not directly linked to epilepsy in everyone, some people with gluten sensitivity or celiac disease may experience neurological symptoms, including seizures. Maintaining a gluten-free diet may be beneficial for those who are sensitive. Occasionally, allergies or sensitivities to specific foods can trigger seizures in susceptible individuals. Some common allergens include dairy, eggs, soy, and nuts[329]. Excessive consumption of caffeine or alcohol might lower the seizure threshold in some individuals. Therefore, moderation or avoidance may be recommended, especially for those who are sensitive to these substances. Poor nutrition or extreme diets lacking in essential nutrients may impact overall health and potentially trigger seizures. As a result, maintaining a well-balanced diet is crucial[330].

It can be helpful to record the foods you eat alongside any seizures you experience. This may help identify patterns or potential triggers and guide dietary adjustments. It's important to discuss any dietary concerns or potential triggers with healthcare professionals, such as neurologists and dietitians, who can provide personalized guidance[331]. If you have epilepsy, it's best to make significant dietary changes gradually so you can monitor any effects on seizure frequency or severity. Also, certain foods or supplements may interact with antiepileptic medications, so it's crucial to discuss any dietary changes or supplements with your healthcare provider to avoid any adverse interactions. Eating a diet rich in fruits, vegetables, whole grains, lean proteins, and healthy fats can contribute to overall health and potentially aid in seizure management[242]. The patient/care giver should carefully track the food intake and its relation to the seizure activity for a period to identify any patterns or correlations. This can help pinpoint specific foods or food groups that might be triggering seizures for the patient. Under medical supervision, the patient can implement elimination and challenge diets to test specific food groups or ingredients as potential triggers. This should be done in a controlled manner to avoid nutritional deficiencies and ensure safety.

Functional nutrients

Functional nutrients are substances found in foods or supplements that provide health benefits beyond basic nutrition. These functional nutrients show promise in potentially aiding seizure control or supporting brain health in individuals with epilepsy. While not a standard treatment for epilepsy, functional nutrients have emerged as promising alternatives or adjuncts to traditional AEDs in managing the condition[9]. These nutrients, thought to go beyond basic nutritional needs and exert specific biological effects, may offer benefits. Certain nutrients, alone or combined with AEDs, have shown potential in reducing seizure activity. Other functional nutrients may support brain health, enhance cognitive function, and protect neurons from damage, especially during seizure activity. Some nutrients can also help alleviate the side effects of certain AEDs, improving overall well-being and quality of life[242].

Omega-3 fatty acids, particularly EPA and DHA, are essential for brain health and function. They have anti-inflammatory properties and might help stabilize neuronal membranes. DHA can be a structural component of neuronal membranes, modulating membrane biophysical properties, ion channel functions, and neurotransmitter signaling[298]. There has been a lot of interest in the potential therapeutic benefits of omega-3 fatty acids for epilepsy, which emerged with the observation that they can reduce cardiac arrhythmia and, consequently, can reduce neuronal excitability[332]. The first clinical study of the therapeutic potential of fish oil in epilepsy patients found that it had an anticonvulsive effect associated with omega-3 fatty acids[333]. However, subsequent clinical studies have produced more controversial results. While some randomized clinical trials have found that consuming approximately 0.6-2 g of fish oil reduces seizure frequency and duration[334], other small-scale, non-randomized studies have reported no regulatory efficacy for seizures[335]. Despite several animal studies suggesting that omega-3 fatty acids suppress seizures, it remains unclear whether they have significant benefits for controlling seizures. Nevertheless, considering the goal of identifying nutritional supplements that assist drug therapy, and given that fish oil is safe at dosages of less than 4 g per day, it can be recommended for regulating seizure generation[336].

Vitamin B6, also known as pyridoxine, has been researched as a potential therapy to treat certain types of epilepsy. It is beneficial in cases of PDE or in conditions that are responsive to pyridoxine supplementation. Vitamin B6 plays a crucial role in synthesizing neurotransmitters, including GABA, which is an inhibitory neurotransmitter that helps regulate neuronal excitability. Vitamin B6 is involved in various metabolic pathways within the nervous system and contributes to overall brain health and function[337]. Individuals with specific metabolic disorders, such as PDE, have shown a positive response to vitamin B6 supplementation. In these cases, the body cannot properly metabolize or utilize vitamin B6, leading to seizures that are responsive to pyridoxine treatment. Although the efficacy of vitamin B6 in treating other forms of epilepsy remains less clear, it may be beneficial in certain cases or when used in combination with other antiepileptic medications. However, more research is needed to establish its widespread effectiveness[56].

It is important to note that responses to vitamin B6 supplementation can vary widely among individuals with epilepsy. Some may experience reduced seizure frequency or severity, while others may not see significant benefits. Vitamin B6 deficiency can negatively impact cognitive function, and supplementation has been associated with improvements in memory, learning, and attention in individuals with epilepsy[338]. Vitamin B6 is also an antioxidant and might protect neurons from damage, potentially contributing to overall brain health and seizure control. Some AEDs can deplete Vitamin B6 Levels, and supplementation can help alleviate associated side effects such as fatigue, irritability, and depression[243]. However, its widespread effectiveness in managing various forms of epilepsy remains an area of ongoing research, and its use as an adjunctive therapy should be guided by healthcare professionals based on individual circumstances.

Vitamin B12, also known as cobalamin, plays a crucial role in various bodily functions, including supporting the health of the nervous system and the synthesis of neurotransmitters[43]. Although research on the specific effects of vitamin B12 on epilepsy is limited, its potential role in maintaining overall nervous system function might have implications for the management of epilepsy. Vitamin B12 is vital for the maintenance of the nervous system. It supports the formation of myelin, which is the protective sheath around nerves, and helps produce neurotransmitters. Vitamin B12, in conjunction with other B vitamins, helps regulate homocysteine levels. High levels of homocysteine have been linked to increased risk of neurological disorders[339]. Vitamin B12's role in supporting nervous system health suggests that it might indirectly influence seizure activity by having neuroprotective effects. Elevated homocysteine levels have been associated with increased susceptibility to seizures[339]. Supplementation with vitamin B12 and other B vitamins might help regulate homocysteine levels and possibly reduce seizure frequency in individuals with high levels[340]. However, direct evidence linking vitamin B12 supplementation to epilepsy management is limited. Studies specifically investigating its efficacy as a treatment for epilepsy are scarce. Fakhroo et al[341] showed that high-dose vitamin B12 pretreatment alleviated the seizure occurrence among pentylenetetrazole-kindled rat models, which suggests that vitamin B12 is a potential strategy to treat epilepsy and other related epileptogenesis activities[341]. It is important to note that responses to vitamin B12 supplementation can vary among individuals. While some may experience improvements in neurological function, including seizure control, others may not see significant benefits. Vitamin B12 is a potential functional nutrient for epilepsy, especially for people who have deficiencies or experience cognitive difficulties or side effects of AEDs. However, due to the limited evidence, its use should be carefully considered and monitored under the guidance of a healthcare professional. Further research is necessary to provide more insights into the possible benefits and risks of using Vitamin B12 for epilepsy management[342].

Vitamin D is a crucial nutrient for maintaining overall health. However, its precise impact on managing epilepsy is still being studied. Lately, it has gained significant attention as a potential functional nutrient for epilepsy management. Its potential implications in epilepsy are of particular interest due to its influence on brain health and immune modulation, although it might indirectly affect neurological health and inflammation regulation. The presence of vitamin D receptors in the brain suggests a role in neuronal health and function. It is involved in neuroprotection, neurogenesis, neuronal differentiation, axonal growth, and the regulation of neurotransmitters[343]. It plays a role in modulating the immune system, reducing inflammation, and supporting overall immune function. Vitamin D's role in supporting neuronal health and protecting against neurodegeneration suggests potential neuroprotective effects[344]. It might indirectly impact seizure susceptibility. Inflammation has been implicated in some types of epilepsy. Vitamin D's anti-inflammatory properties might help regulate immune responses that could affect seizure activity[93]. Research specifically focusing on vitamin D as a treatment for epilepsy is limited. While some observational studies suggest an association between low vitamin D levels and increased seizure frequency[84], direct causation remains unclear. Low levels of vitamin D have been observed in some individuals with epilepsy. Holló‎ et al[97] found that correcting vitamin D deficiency helped improve seizure control. Therefore, supplementation might be considered for those with deficiencies, but its direct impact on seizure control requires further investigation.

Vitamin E, a fat-soluble antioxidant, is known for its role in protecting cells from oxidative damage. Its potential implications in epilepsy have been explored due to its antioxidant properties and influence on neurological health[345]. Vitamin E is an antioxidant, scavenging free radicals and reducing cell oxidative stress. This function helps protect cell membranes from damage. Its antioxidant properties suggest potential neuroprotective effects, which might affect brain health and neuronal function[346]. Oxidative stress and neuronal damage are implicated in certain types of epilepsy. Vitamin E's antioxidant properties might contribute to protecting neurons from damage, potentially influencing seizure activity. Vitamin E's anti-inflammatory properties might help modulate inflammatory responses in the brain, which could affect seizure susceptibility[347]. Studies specifically investigating vitamin E as a treatment for epilepsy are limited. While some research suggests a potential role in neuroprotection, its direct impact on seizure control remains unclear. Ogunmekan et al[348] showed that daily supplementation with 400 IU of vitamin E for three months reduces seizure frequency by 50% in patients with epilepsy[348]. On the other hand, Raju et al[349] showed no significant change in seizure frequency after adding vitamin E to AEDs in patients with uncontrolled epilepsy. Responses to vitamin E supplementation can vary among individuals. Some might experience benefits in overall health, including potential neuronal protection, while others might not see significant effects. Vitamin E supplementation might be considered for individuals with epilepsy, especially if there are indications of increased oxidative stress or a deficiency[105]. However, its specific impact on seizures requires more research. On the other hand, high doses of Vitamin E can increase the risk of bleeding and potentially interact with certain medications[350].

Vitamin C, or ascorbic acid, is an antioxidant that dissolves in water and has various bodily roles. These roles include enhancing immune function, collagen synthesis, and antioxidant activity[340]. Concerning epilepsy, its antioxidant properties and influence on overall health are particularly relevant[345]. Vitamin C is an antioxidant by scavenging free radicals and reducing cell oxidative stress. This helps to protect cells from damage caused by ROS[351]. Vitamin C is essential for producing collagen, which is vital for connective tissues, wound healing, and maintaining the integrity of blood vessels[352]. Furthermore, vitamin C is a coenzyme for the enzymatic conversion of dopamine to norepinephrine, which helps regulate neurotransmission[353]. Oxidative stress is implicated in certain types of epilepsy, and vitamin C's antioxidant properties might contribute to protecting neurons from oxidative damage, potentially impacting seizure activity[27]. Although not directly studied in epilepsy, vitamin C's neuroprotective potential might affect overall brain health and neuronal function. While there are limited studies on vitamin C as a treatment for epilepsy, some suggest that Vitamin C supplementation may decrease seizure frequency and severity in individuals with epilepsy, particularly those with low levels[21]. The neuroprotective effect and the anti-inflammatory properties of vitamin C might contribute to improved seizure control and overall brain health. Additionally, vitamin C can enhance the permeability of the BBB, allowing for better transport of essential nutrients and medications into the brain[354]. This could potentially enhance the effectiveness of AED. Although its antioxidant properties are recognized, the direct impact of vitamin C on seizure control remains unclear and requires further investigation. Vitamin C is essential for overall health, immune function, and collagen synthesis. Ensuring adequate levels might indirectly support the body's ability to manage various health conditions, including epilepsy[81]. While vitamin C supplementation is generally safe, its specific impact on seizures needs further research. It might be considered as part of a well-balanced diet to support overall health, including potential neuroprotective effects.

L-carnitine is a compound derived from an amino acid that helps in energy metabolism by transporting fatty acids into mitochondria to produce energy[355]. It has been studied for its potential role in managing epilepsy due to its neuroprotective properties and role in energy metabolism. L-carnitine is essential in facilitating the transport of fatty acids to mitochondria, where they are metabolized to produce energy. It is essential in tissues that rely on fatty acids for energy production[356]. L-carnitine has antioxidant properties, which makes it useful in protecting against neuronal damage and reducing oxidative stress. Some types of epilepsy are linked to abnormalities in brain energy metabolism. L-carnitine's role in fatty acid transport and energy production might affect brain energy metabolism and neuronal function. Its antioxidant properties suggest potential neuroprotective effects that might indirectly influence seizure activity by protecting neurons from damage[357].

L-carnitine requirements vary based on dietary intake, absorption, and underlying medical conditions. Research on L-carnitine as a treatment for epilepsy is limited, preliminary, and mainly focused on specific groups, such as children with mitochondrial disorders[179]. It is necessary to conduct more extensive research to confirm its benefits and identify individual responses. Some studies suggest that L-carnitine supplementation can help decrease seizure frequency and severity, particularly in individuals with deficiencies. L-carnitine deficiency can negatively impact cognitive function. Hussein et al[356] showed that four weeks of L-carnitine supplementation (100 mg/kg/day) was associated with significantly reduced seizure score, observed as early as the second day of treatment and continued during the treatment phase. Supplementation has been found to improve memory, learning, and attention in individuals with epilepsy. Additionally, certain anti-epileptic drugs can deplete L-carnitine levels, leading to side effects like fatigue, muscle weakness, and depression. Supplementation might help alleviate such side effects. Carnitine supplementation maintains constant FC serum levels in patients treated with AEDs such as VPA. Carnitine supplementation may revert the latent pancreatic injury induced by VPA as indicated by decreasing serum amylase levels[184]. L-carnitine supplementation might be considered for individuals with epilepsy, particularly in cases where there are indications of deficiencies or metabolic abnormalities related to fatty acid metabolism[187]. It is important to note that L-carnitine should not replace conventional anti-epileptic drugs but can be used as a complementary therapy to optimize seizure control and overall health[358].

Pyruvate is a crucial component in the body's energy production, showing promise as a functional nutrient in treating epilepsy. This adaptable molecule has potential in multiple facets of seizure management, making it an exciting area of research[359]. Its role in glucose metabolism is essential to its efficacy, as it is an intermediary in the breakdown of glucose for energy. It typically enters the Krebs cycle, a cellular powerhouse producing ATP, essential fuel for bodily functions, including brain activity[360]. Pyruvate also acts as an antioxidant, protecting brain cells from damage caused by harmful free radicals like hydrogen peroxide, potentially reducing seizure risks[361]. Inflammation can be significant in epilepsy, but ethyl pyruvate, a derivative, has impressive anti-inflammatory capabilities, hinting at its potential to control seizures[362]. Encouraging animal studies have shown that oral pyruvate administration significantly decreases seizure frequency, indicating therapeutic promise. Popova et al[363] showed that chronic oral pyruvate intake succeeded in completely abolishing seizures in three different established epileptic phenotypes of acquired epilepsy models in rodents]. Combining pyruvate with antioxidants such as vitamins C and E could augment its anti-seizure effects[364]. While clinical trials are necessary to gauge its therapeutic efficacy and viability as a treatment for acquired epilepsies in humans, pyruvate's multifaceted characteristics position it as a captivating contender for epilepsy management. Its ability to address various seizure-related factors, from energy production to inflammation control, presents a unique and potentially impactful approach to enhancing the lives of individuals living with epilepsy[365].

Coenzyme Q10 (CoQ10) is a compound naturally produced by the body and serves as an antioxidant, aiding in cell energy production. Although research on CoQ10 specifically targeting epilepsy is limited, its role in cellular energy metabolism and antioxidant properties suggest potential implications for managing seizures[366]. CoQ10 plays a crucial role in the mitochondria, where it helps generate ATP, the body's primary energy source. CoQ10 acts as an antioxidant, scavenging free radicals and reducing oxidative stress that can damage cells[367]. Some types of epilepsy involve mitochondrial dysfunction and impaired energy metabolism. CoQ10's role in mitochondrial function might have implications for supporting cellular energy production in the brain. CoQ10's antioxidant properties suggest potential neuroprotective effects, which might indirectly influence seizure activity by protecting neurons from oxidative damage[368]. Research specifically focusing on CoQ10 as a treatment for epilepsy is limited. Tawfik et al[369] showed that CoQ10 can potentially reduce seizure severity and oxidative stress induced by pilocarpine in rats. CoQ10 is a safe and effective adjuvant to phenytoin therapy in epilepsy, enhancing its antiepileptic effects and protecting against seizure-induced cognitive impairment and oxidative damage caused by chronic phenytoin use[369]. While its role in mitochondrial function and antioxidative properties is recognized, its direct impact on seizure control remains unclear and needs further investigation. Responses to CoQ10 supplementation can vary among individuals. Some might experience improvements in overall health, including potential neurological benefits, while others might not see significant effects. CoQ10 supplementation might be considered for individuals with epilepsy, especially in cases where there are indications of mitochondrial dysfunction or increased oxidative stress[370].

Curcumin (diferuloylmethane) is a turmeric compound known for its potential health benefits, including anti-inflammatory and antioxidant properties. While research on curcumin's effects on epilepsy is still in its early stages, it has been shown to have neuroprotective effects that could help manage seizures[371]. Curcumin's anti-inflammatory properties can reduce inflammation-related molecular pathways that contribute to certain types of epilepsy[372]. Additionally, its antioxidant properties can help protect neurons from oxidative damage, which may indirectly affect seizure susceptibility. However, further research is needed to understand curcumin's direct impact on seizure control[373]. Studies suggest that curcumin may alleviate epileptic seizures by modulating oxidative stress, neuroinflammation, and neurotransmission while also inhibiting apoptotic cell death and necrotic and autophagic pathways in hippocampal neurons[374-376]. Furthermore, curcumin has been found to have a protective effect on relieving memory impairment by affecting brain monoamine levels[377].

Numerous animal studies have shown that Curcumin can be an effective adjuvant therapy for epilepsy. For instance, in a study by Mehla et al[378], it was found that pretreatment with curcumin improved seizures, oxidative stress, and cognitive impairment in rats with pentylenetetrazol-induced kindling[378]. Similarly, Agarwal‎ et al[379] found that liposome-entrapped curcumin (in doses of 25 and 50 mg/kg) resulted in a significant increase in seizure threshold current and latency to myoclonic and generalized seizures in increasing current electroshock seizures test and pentylenetetrazol-induced seizures, respectively[379]. Another study by Jiang demonstrated the protective effect of curcumin against cognitive impairment and its ability to modify epileptogenesis in a post-status epilepticus temporal lobe epilepsy model[380]. However, clinical trials on humans with epilepsy are still needed to confirm the efficacy of curcumin.

The effectiveness of curcumin as a therapeutic agent is limited by its poor bioavailability in the human body. Researchers are currently exploring various formulations and strategies to improve its absorption and efficacy[381]. However, curcumin supplementation may be considered as a potential therapeutic option for individuals suffering from epilepsy, especially in cases where inflammation or oxidative stress may be contributing factors[382]. Although curcumin has anti-inflammatory and antioxidant properties, further investigation is needed to establish its specific impact on managing epilepsy. Curcumin's ability to modulate inflammation and protect neurons may offer new avenues for supporting seizure control, but more research, particularly clinical trials, is required to determine its efficacy and safety as a treatment for epilepsy[383]. Therefore, it is essential to approach curcumin supplementation for epilepsy under professional guidance and with utmost caution.

Magnesium is an important mineral found inside our body's cells. It acts as a helper in over 300 chemical reactions in our body and plays a crucial role in various bodily functions, including nerve function and muscle control. Magnesium has emerged as a potential therapy for managing epilepsy. This is because it helps regulate neurotransmitters, and its ability to modulate neuronal excitability has led researchers to explore its potential benefits in managing epilepsy[384]. Magnesium acts as a neuroprotective agent, which potentially protects against the damage caused by excessive neuronal firing during seizures. It also helps regulate NMDA receptors, which control neurotransmitters' activity in the brain. By modulating these receptors, magnesium might help prevent excessive neuronal excitability[113].

Magnesium deficiency is associated with increased neuronal excitability, which is a known precursor to seizure activity. Its supplementation has been shown to stabilize neuronal membranes, potentially reducing seizure frequency and severity in individuals with suboptimal magnesium levels[121]. Like some conventional AEDs, magnesium is a natural NMDA receptor antagonist and non-selective calcium channel blocker. This modulatory effect can directly suppress excessive neuronal firing, a hallmark of epileptic seizures[374]. Magnesium‎ also is a cofactor for GABA synthesis, accounting for its anti-seizure activity[123].

Moreover, magnesium possesses potent antioxidant properties, scavenging free radicals and protecting neurons from excitotoxic damage[385,386]. This neuroprotective effect can mitigate seizure-induced neuronal injury and contribute to long-term brain health in individuals with epilepsy. Magnesium is also crucial in maintaining mitochondrial integrity and promoting efficient energy production within brain cells[387]. By supporting mitochondrial function, magnesium can prevent energy imbalances contributing to seizure initiation and propagation. As epilepsy is often accompanied by mood and cognitive impairments, magnesium's neuroprotective and anti-inflammatory properties may exert beneficial effects on mood regulation[388], memory, and concentration[389], potentially improving overall well-being in individuals with epilepsy.

Research suggests that taking magnesium supplements may help reduce the frequency and severity of seizures in individuals with epilepsy. Many animal studies have shown that magnesium supplements can effectively reduce seizure frequency and severity. Safar et al[390] found that magnesium supplements can enhance the effectiveness of AED, such as VPA, by improving redox balance and modulating certain brain amino acids. Clinical trials have also shown promising results, with magnesium supplements leading to moderate to significant anticonvulsant effects in patients with different types of epilepsy. Abdelmalik et al[124] reported significant improvements in seizure control and frequency in patients with drug-resistant seizures who took oral magnesium oxide (420 mg twice a day), with some becoming seizure-free[112]. While Yadav et al[391] did not find a significant difference in seizure control or neurodevelopmental outcomes, they did find that EEG readings improved in children with West syndrome who received magnesium sulphate[391]. Additionally, intravenous infusion of magnesium sulphate remains the drug of choice to prevent and control convulsions caused by eclampsia, significantly reducing maternal mortality related to eclampsia[392].

While magnesium is not typically used as a standalone treatment for epilepsy, it may serve as an adjunctive therapy alongside standard antiepileptic medications. Combining magnesium supplementation with existing treatments might enhance their efficacy or reduce specific side effects[393]. Maintaining a balanced diet that includes magnesium-rich foods such as almonds, nuts (e.g., cashews), oatmeal, seeds (pumpkin, sunflowers, sesame), black beans, brown rice, flaxseed, leafy green vegetables (spinach), broccoli, bananas, soybeans, sweet corn, whole grains, and certain types of fish is essential[394]. However, if magnesium supplementation is recommended, it should be done under medical supervision to ensure safety and appropriate dosage. Further research is warranted to optimize magnesium dosing strategies and elucidate ‎its precise mechanisms of action in epileptic seizures. Additionally, long-term studies ‎are needed to assess the potential benefits of magnesium supplementation for mood, ‎cognitive function, and overall quality of life in individuals with epilepsy.‎

Medium-chain fatty acids (MCFAs) have garnered attention for their potential in reducing epilepsy through various mechanisms. MCFAs are metabolized differently than long-chain fatty acids, rapidly converting to ketone bodies in the liver, such as beta-hydroxybutyrate and acetoacetate, which can serve as alternative fuel sources for the brain during metabolic stress[395]. Additionally, MCFAs may enhance GABA production, the brain's primary inhibitory neurotransmitter, thereby reducing neuronal excitability. Their anti-inflammatory properties may mitigate neuroinflammation, while improvements in mitochondrial function and modulation of ion channels contribute to stabilizing neuronal membranes and reducing hyperexcitability[396]. Collectively, these mechanisms suggest that MCFAs hold promise as adjunctive therapy for epilepsy management, although further research is warranted to fully elucidate their efficacy and underlying mechanisms.

Incorporating foods rich in MCFAs into one's diet involves sourcing from various natural sources. Coconut oil emerges as a prominent option due to its high content of MCFAs, notably lauric acid, suitable for culinary applications such as cooking, baking, or inclusion in beverages and dressings. Likewise, coconut-derived products like coconut milk, cream, and shredded coconut provide convenient alternatives, lending themselves well to diverse recipes[397]. Palm kernel oil, sharing similarities with coconut oil, also serves as a source of MCFAs for cooking or baking purposes[398]. Dairy products, including butter, cheese, and full-fat yogurt, albeit containing modest amounts of MCFAs, offer additional dietary options, particularly when sourced from grass-fed animals[399]. Alternatively, individuals seeking concentrated MCFAs can explore mediums like MCT oil or MCT powder, derived from coconut or palm kernel oil, easily incorporated into beverages or recipes[400]. Maintaining dietary balance is paramount, ensuring MCFAs complement a spectrum of nutrient-rich foods for holistic health benefits. As with any dietary modifications, seeking guidance from healthcare professionals is advisable, especially concerning specific dietary needs or medical conditions.

General dietary considerations

Ensuring adequate intake of essential macronutrients (carbohydrates, proteins, fats) and micronutrients (vitamins, minerals) is paramount for optimal brain health and energy metabolism, both crucial for seizure control. Balanced nutritional planning, potentially with the support of a registered dietitian, is recommended. Controlled fasting or intermittent fasting regimens have shown promise in reducing seizure frequency, possibly due to the metabolic shift towards ketone utilization during fasting periods[401]. However, patients with epilepsy should avoid blood sugar fluctuations. Patients with epilepsy may prioritize low-glycemic index foods and limit refined carbohydrates and sugary ‎drinks[402].‎ Adequate hydration is vital for various physiological processes, including brain function. Dehydration can exacerbate seizure activity, making consistent water intake essential for individuals with epilepsy[147]. Dietary needs and responses vary depending on age, seizure type, underlying medical conditions, and individual preferences. A personalized approach tailored to each patient's needs and implemented under the guidance of a healthcare professional is crucial for ensuring safety and maximizing potential benefits. Dietary modifications may impact antiepileptic medications' efficacy or side effects, requiring close monitoring and potential adjustments[223]. In addition, some AEDs can deplete electrolytes like sodium, potassium, ‎magnesium, and calcium. Ensuring adequate intake of these electrolytes through your diet is ‎important[403].‎ It's crucial to implement any dietary changes under the guidance of healthcare providers, ‎particularly a registered dietitian or a healthcare team specialized in epilepsy management. ‎Monitoring nutritional intake, regular check-ups, and tailored dietary plans are essential to ‎ensure that nutritional needs are met while managing epilepsy effectively. Additionally, ‎these dietary changes might interact with medications, so medical supervision is crucial for ‎safety and efficacy[13].‎ Maintaining a healthy lifestyle with regular exercise and adequate sleep is essential to ‎complement the nutritional and medication management plan[404].‎

Challenges of implementing nutritional interventions in clinical settings

Implementing nutritional interventions in epilepsy management presents several significant challenges that must be carefully navigated to ensure efficacy and patient safety.

Patient compliance and individual variability: One of the foremost challenges is ensuring patient compliance with prescribed dietary regimens, particularly when these diets require significant lifestyle changes, such as the ketogenic diet. Strict adherence is critical for effectiveness, yet many patients may find it challenging to maintain such diets over the long term due to the restrictive nature of the interventions[405]. Additionally, epilepsy is a highly heterogeneous condition, with substantial variability in seizure types, underlying causes, and patient responses to treatments. This variability extends to nutritional needs, where what is effective for one individual may not be suitable for another. Developing personalized nutrition plans tailored to each patient’s unique metabolic profile, genetic predispositions, and personal circumstances is ideal but challenging, often requiring advanced diagnostic tools and ongoing adjustments, which can be resource-intensive[406].

Resource allocation and healthcare system support: Implementing and sustaining nutritional interventions also demands considerable resources, including access to specialized dietitians, frequent monitoring, and comprehensive patient education. In many clinical settings, particularly those with limited resources, the availability of such specialized care may be constrained. Furthermore, the healthcare system’s infrastructure must support the integration of nutrition into epilepsy care, which may require additional training for healthcare providers, interprofessional collaboration, and the development of clear protocols to manage these interventions effectively[407].

Monitoring and adjusting dietary plans: Ongoing monitoring is essential to ensure that nutritional interventions are both effective and safe, requiring regular assessments of patients' dietary adherence, metabolic status, and seizure control. This dynamic process necessitates close collaboration between patients and healthcare providers, with potential challenges arising from the need for frequent laboratory tests, dietary assessments, and the adjustment of dietary plans based on clinical outcomes[408]. The burden of continuous monitoring and adjustment can be significant, both for patients, who must remain engaged and compliant, and for healthcare providers, who must allocate time and resources to these activities[409].

Psychosocial and cultural considerations: Nutritional interventions can profoundly impact a patient's social and cultural life. Strict dietary regimens may limit participation in social activities or conflict with cultural dietary practices, leading to social isolation or emotional distress. The psychological burden of adhering to such diets, especially in the context of a chronic condition like epilepsy, can also lead to mental health challenges, including anxiety, depression, or disordered eating behaviors. Addressing these psychosocial factors requires a holistic approach that includes psychological support and consideration of the patient’s cultural background[410,411].

Interaction with AED: Nutritional interventions can interact with AED, potentially affecting drug efficacy or increasing the risk of side effects. For example, certain diets may alter the metabolism or absorption of these drugs, leading to subtherapeutic levels or toxicity. Managing these interactions requires careful coordination among dietitians, pharmacists, and neurologists to avoid compromising the effectiveness of antiepileptic therapy, particularly in patients on multiple medications[412].

Educational and training needs: Finally, the successful implementation of nutritional interventions in epilepsy care necessitates adequate training and education for healthcare providers. The evolving nature of research in this field requires that clinicians remain informed about the latest evidence and best practices. In many settings, a lack of specialized training may hinder the ability to deliver effective nutritional care, underscoring the need for ongoing professional development and education[413].

Novel insights, therapeutic implications, and future research directions

Our review reveals that nutrient deficiencies—such as those in vitamins B6, folate, and D—disrupt neurotransmitter synthesis, compromise myelin integrity, and contribute significantly to oxidative stress and neuroinflammation. These processes lower the seizure threshold, thereby increasing susceptibility to epileptogenesis[414]. Moreover, emerging evidence indicates that malnutrition-induced alterations in the gut microbiota may compromise the gut-brain axis, further exacerbating neuroinflammatory responses and influencing neuronal excitability[415]. Together, these interrelated pathways underscore a multifaceted, bidirectional link in which malnutrition may both predispose individuals to epilepsy and result from the metabolic and pharmacological challenges associated with its management.

Building on these mechanistic insights, recent therapeutic approaches have begun to explore nutritional interventions as adjuncts to conventional antiepileptic treatments. Dietary strategies—such as the ketogenic diet, modified Atkins diet, and low glycemic index diet—have shown promise in reducing seizure frequency, potentially by enhancing mitochondrial function, mitigating oxidative stress, and modulating inflammatory pathways[297]. Additionally, targeted micronutrient supplementation (e.g., vitamin D, magnesium, omega-3 fatty acids) may help restore neuronal stability by correcting specific deficiencies contributing to dysregulated neurotransmission[416].

Despite these promising avenues, significant gaps remain in our understanding of how nutritional factors modulate epileptogenesis. Future research should prioritize:

Mechanistic studies: Investigating how specific nutrient deficiencies affect neuronal excitability and elucidating the precise role of gut microbiota alterations in mediating neuroinflammatory processes.

Clinical trials: Conducting long-term, well-controlled studies to evaluate personalized nutritional interventions that combine micronutrient repletion with strategies to modulate the gut microbiota, thereby determining their efficacy in reducing seizure frequency and severity.

Biomarker development: Identifying nutritional and microbial biomarkers that predict seizure susceptibility and therapeutic response would facilitate the development of targeted, individualized treatment protocols.

Recommendations

Based on the multifaceted interplay between nutrition and epilepsy highlighted in this review, several recommendations emerge. Healthcare providers should prioritize comprehensive nutritional assessments for individuals with epilepsy, acknowledging the bidirectional impact between the condition and nutritional status. Tailored dietary plans, guided by registered dietitians or nutrition specialists, should be integrated into the holistic management of epilepsy, keeping in mind the unique characteristics and needs of each individual with epilepsy. Furthermore, research efforts should focus on elucidating specific nutritional deficiencies or imbalances that exacerbate seizure susceptibility, thereby fostering targeted interventions. Clinical trials evaluating the efficacy of dietary modifications, supplements, or personalized nutrition plans in seizure control are imperative. Additionally, public health initiatives advocating for awareness about the role of nutrition in epilepsy care and fostering access to nutritional support services are essential for optimizing patient outcomes and enhancing overall well-being.

Limitations

Despite the comprehensive analysis presented in this review, several limitations warrant acknowledgment. A key challenge is the heterogeneity in study designs, participant characteristics, diagnostic criteria, intervention protocols, and follow-up durations across the included studies. This variability makes direct comparisons difficult and contributes to inconsistencies in reported outcomes. Differences in epilepsy classification, seizure severity, dietary interventions, and assessment tools further complicate the synthesis of findings, impacting the generalizability of results. Additionally, most of the included studies are observational or based on animal models, with a relative scarcity of RCTs and long-term prospective studies. Observational studies are inherently susceptible to confounding factors, making it difficult to establish causality between nutritional factors and epilepsy. While animal models provide mechanistic insights, their applicability to human physiology remains limited. These methodological constraints restrict the strength of the evidence and underscore the need for well-designed RCTs to validate the role of nutritional interventions in epilepsy management. Furthermore, the complex interplay between nutrition and epilepsy—involving neurotransmitter regulation, neuronal plasticity, oxidative stress, and gut microbiota interactions—remains incompletely elucidated. Many mechanisms remain speculative, requiring further mechanistic and clinical research. The review also acknowledges potential limitations in literature retrieval, as database coverage, language restrictions, and publication bias may have led to the omission of relevant studies. The tendency for studies with positive findings to be published more frequently than those with negative or inconclusive results may further influence the comprehensiveness and objectivity of the conclusions. Finally, individual variability in response to dietary interventions and supplements complicates the generalization of recommendations. Factors such as genetic predisposition, comorbidities, medication interactions, and dietary adherence contribute to differences in treatment outcomes. Given these limitations, future research should prioritize methodological consistency, longer follow-up durations, and diverse population cohorts to enhance the reliability and clinical applicability of findings in the field of epilepsy and nutrition.

CONCLUSION

This comprehensive review illuminates the intricate nexus between malnutrition and epilepsy, showcasing their bidirectional influence on each other. The exploration of malnutrition as a potential risk factor for epilepsy underscores the significance of adequate nutrition in reducing neurological vulnerabilities. Similarly, the analysis of how epilepsy impacts nutritional status emphasizes the complexities of altered dietary patterns and metabolic perturbations in individuals with epilepsy. Moreover, the insights gleaned from exploring nutritional management strategies unveil promising avenues for mitigating seizure susceptibility and enhancing overall brain health. This review underscores the imperative of a holistic approach, integrating nutrition into epilepsy management strategies, offering a roadmap for future research endeavors and personalized therapeutic interventions in epilepsy care.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: Bahrain

Peer-review report’s classification

Scientific Quality: Grade B, Grade B, Grade C, Grade D

Novelty: Grade B, Grade B, Grade C, Grade C

Creativity or Innovation: Grade B, Grade B, Grade C, Grade C

Scientific Significance: Grade A, Grade B, Grade C, Grade D

P-Reviewer: Chhetri KB; Kandasamy M; Ma JY S-Editor: Liu H L-Editor: A P-Editor: Guo X

References
1.  Stafstrom CE, Carmant L. Seizures and epilepsy: an overview for neuroscientists. Cold Spring Harb Perspect Med. 2015;5:a022426.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in RCA: 440]  [Article Influence: 44.0]  [Reference Citation Analysis (0)]
2.  Russ SA, Larson K, Halfon N. A national profile of childhood epilepsy and seizure disorder. Pediatrics. 2012;129:256-264.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 387]  [Cited by in RCA: 399]  [Article Influence: 30.7]  [Reference Citation Analysis (0)]
3.  Beghi E, Hesdorffer D. Prevalence of epilepsy--an unknown quantity. Epilepsia. 2014;55:963-967.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in RCA: 50]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
4.  Zack MM, Kobau R. National and State Estimates of the Numbers of Adults and Children with Active Epilepsy - United States, 2015. MMWR Morb Mortal Wkly Rep. 2017;66:821-825.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in RCA: 425]  [Article Influence: 53.1]  [Reference Citation Analysis (0)]
5.  Sirven JI. Epilepsy: A Spectrum Disorder. Cold Spring Harb Perspect Med. 2015;5:a022848.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in RCA: 105]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
6.  Falco-Walter J. Epilepsy-Definition, Classification, Pathophysiology, and Epidemiology. Semin Neurol. 2020;40:617-623.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 148]  [Article Influence: 29.6]  [Reference Citation Analysis (0)]
7.  Pack AM. Epilepsy Overview and Revised Classification of Seizures and Epilepsies. Continuum (Minneap Minn). 2019;25:306-321.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 36]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
8.  Jette N, Reid AY, Wiebe S. Surgical management of epilepsy. CMAJ. 2014;186:997-1004.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
9.  Kiani AK, Dhuli K, Donato K, Aquilanti B, Velluti V, Matera G, Iaconelli A, Connelly ST, Bellinato F, Gisondi P, Bertelli M. Main nutritional deficiencies. J Prev Med Hyg. 2022;63:E93-E101.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
10.  Jahromi SR, Togha M, Fesharaki SH, Najafi M, Moghadam NB, Kheradmand JA, Kazemi H, Gorji A. Gastrointestinal adverse effects of antiepileptic drugs in intractable epileptic patients. Seizure. 2011;20:343-346.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in RCA: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
11.  Siddiqui F, Salam RA, Lassi ZS, Das JK. The Intertwined Relationship Between Malnutrition and Poverty. Front Public Health. 2020;8:453.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in RCA: 108]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
12.  Guarnieri L, Bosco F, Ruga S, Tallarico M, Leo A, De Sarro G, Citraro R. Long-Term High-Fat Diet Aggravates Absence Seizures and Neurobehavioral Disorders Without Inducing Metabolic Disorders in WAG/Rij Rats: Involvement of Systemic and Central Inflammation. Mol Neurobiol. 2024;62:4837-4853.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
13.  Soltani D, Ghaffar Pour M, Tafakhori A, Sarraf P, Bitarafan S. Nutritional Aspects of Treatment in Epileptic Patients. Iran J Child Neurol. 2016;10:1-12.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Tan BL, Norhaizan ME. Effect of High-Fat Diets on Oxidative Stress, Cellular Inflammatory Response and Cognitive Function. Nutrients. 2019;11:2579.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in RCA: 263]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
15.  Kurowska A, Ziemichód W, Herbet M, Piątkowska-Chmiel I. The Role of Diet as a Modulator of the Inflammatory Process in the Neurological Diseases. Nutrients. 2023;15:1436.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 39]  [Reference Citation Analysis (0)]
16.  Zielińska M, Łuszczki E, Dereń K. Dietary Nutrient Deficiencies and Risk of Depression (Review Article 2018-2023). Nutrients. 2023;15:2433.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
17.  Kirkland AE, Sarlo GL, Holton KF. The Role of Magnesium in Neurological Disorders. Nutrients. 2018;10:730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in RCA: 182]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
18.  Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr. 2021;12:1239-1285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in RCA: 153]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
19.  Pietrzak D, Kasperek K, Rękawek P, Piątkowska-Chmiel I. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients. 2022;14:1952.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 46]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
20.  Sharma S, Jain P. The ketogenic diet and other dietary treatments for refractory epilepsy in children. Ann Indian Acad Neurol. 2014;17:253-258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
21.  Szałwińska K, Cyuńczyk M, Kochanowicz J, Witkowska AM. Dietary and lifestyle behavior in adults with epilepsy needs improvement: a case-control study from northeastern Poland. Nutr J. 2021;20:62.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
22.  Kim JE, Cho KO. Functional Nutrients for Epilepsy. Nutrients. 2019;11:1309.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
23.  Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, Squadrito F, Altavilla D, Bitto A. Oxidative Stress: Harms and Benefits for Human Health. Oxid Med Cell Longev. 2017;2017:8416763.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1959]  [Cited by in RCA: 2175]  [Article Influence: 271.9]  [Reference Citation Analysis (0)]
24.  Chowanadisai W, Kelleher SL, Lönnerdal B. Maternal zinc deficiency reduces NMDA receptor expression in neonatal rat brain, which persists into early adulthood. J Neurochem. 2005;94:510-519.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in RCA: 53]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
25.  Al-Beltagi M, Saeed NK. Epilepsy and the gut: Perpetrator or victim? World J Gastrointest Pathophysiol. 2022;13:143-156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 2]  [Cited by in RCA: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (4)]
26.  Sumadewi KT, Harkitasari S, Tjandra DC. Biomolecular mechanisms of epileptic seizures and epilepsy: a review. Acta Epileptologica. 2023;5:28.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
27.  Borowicz-Reutt KK, Czuczwar SJ. Role of oxidative stress in epileptogenesis and potential implications for therapy. Pharmacol Rep. 2020;72:1218-1226.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 97]  [Article Influence: 19.4]  [Reference Citation Analysis (0)]
28.  Madireddy S, Madireddy S. Therapeutic Strategies to Ameliorate Neuronal Damage in Epilepsy by Regulating Oxidative Stress, Mitochondrial Dysfunction, and Neuroinflammation. Brain Sci. 2023;13:784.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 21]  [Reference Citation Analysis (0)]
29.  Dawson MI. The importance of vitamin A in nutrition. Curr Pharm Des. 2000;6:311-325.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in RCA: 73]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
30.  Maden M, Gale E, Zile M. The role of vitamin A in the development of the central nervous system. J Nutr. 1998;128:471S-475S.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in RCA: 86]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
31.  Olson CR, Mello CV. Significance of vitamin A to brain function, behavior and learning. Mol Nutr Food Res. 2010;54:489-495.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in RCA: 82]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
32.  Ho YH, Lin YT, Wu CW, Chao YM, Chang AY, Chan JY. Peripheral inflammation increases seizure susceptibility via the induction of neuroinflammation and oxidative stress in the hippocampus. J Biomed Sci. 2015;22:46.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in RCA: 98]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
33.  Etchamendy N, Enderlin V, Marighetto A, Vouimba RM, Pallet V, Jaffard R, Higueret P. Alleviation of a selective age-related relational memory deficit in mice by pharmacologically induced normalization of brain retinoid signaling. J Neurosci. 2001;21:6423-6429.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in RCA: 150]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
34.  Misner DL, Jacobs S, Shimizu Y, de Urquiza AM, Solomin L, Perlmann T, De Luca LM, Stevens CF, Evans RM. Vitamin A deprivation results in reversible loss of hippocampal long-term synaptic plasticity. Proc Natl Acad Sci U S A. 2001;98:11714-11719.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in RCA: 190]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
35.  Etchamendy N, Enderlin V, Marighetto A, Pallet V, Higueret P, Jaffard R. Vitamin A deficiency and relational memory deficit in adult mice: relationships with changes in brain retinoid signalling. Behav Brain Res. 2003;145:37-49.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in RCA: 142]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
36.  Sayyah M, Yousefi-Pour M, Narenjkar J. Anti-epileptogenic effect of beta-carotene and vitamin A in pentylenetetrazole-kindling model of epilepsy in mice. Epilepsy Res. 2005;63:11-16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
37.  Rosiles-Abonce A, Rubio C, Taddei E, Rosiles D, Rubio-Osornio M. Antiepileptogenic Effect of Retinoic Acid. Curr Neuropharmacol. 2021;19:383-391.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
38.  Smith TJ, Johnson CR, Koshy R, Hess SY, Qureshi UA, Mynak ML, Fischer PR. Thiamine deficiency disorders: a clinical perspective. Ann N Y Acad Sci. 2021;1498:9-28.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 89]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
39.  Mrowicka M, Mrowicki J, Dragan G, Majsterek I. The importance of thiamine (vitamin B1) in humans. Biosci Rep. 2023;43.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
40.  Yu Q, Liu H, Sang S, Chen L, Zhao Y, Wang Y, Zhong C. Thiamine deficiency contributes to synapse and neural circuit defects. Biol Res. 2018;51:35.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
41.  Dhir S, Tarasenko M, Napoli E, Giulivi C. Neurological, Psychiatric, and Biochemical Aspects of Thiamine Deficiency in Children and Adults. Front Psychiatry. 2019;10:207.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in RCA: 122]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
42.  Abdou E, Hazell AS. Thiamine deficiency: an update of pathophysiologic mechanisms and future therapeutic considerations. Neurochem Res. 2015;40:353-361.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in RCA: 70]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
43.  Calderón-Ospina CA, Nava-Mesa MO. B Vitamins in the nervous system: Current knowledge of the biochemical modes of action and synergies of thiamine, pyridoxine, and cobalamin. CNS Neurosci Ther. 2020;26:5-13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in RCA: 231]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
44.  Sechi G, Serra A. Wernicke's encephalopathy: new clinical settings and recent advances in diagnosis and management. Lancet Neurol. 2007;6:442-455.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 729]  [Cited by in RCA: 722]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
45.  Pawar RD, Balaji L, Grossestreuer AV, Thompson G, Holmberg MJ, Issa MS, Patel PV, Kronen R, Berg KM, Moskowitz A, Donnino MW. Thiamine Supplementation in Patients With Alcohol Use Disorder Presenting With Acute Critical Illness : A Nationwide Retrospective Observational Study. Ann Intern Med. 2022;175:191-197.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
46.  Pinto JT, Zempleni J. Riboflavin. Adv Nutr. 2016;7:973-975.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in RCA: 95]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
47.  Holton KF. Micronutrients May Be a Unique Weapon Against the Neurotoxic Triad of Excitotoxicity, Oxidative Stress and Neuroinflammation: A Perspective. Front Neurosci. 2021;15:726457.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
48.  Kmoch S, Zeman J, Hrebícek M, Ryba L, Kristensen MJ, Gregersen N. Riboflavin-responsive epilepsy in a patient with SER209 variant form of short-chain acyl-CoA dehydrogenase. J Inherit Metab Dis. 1995;18:227-229.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 15]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
49.  Udhayabanu T, Manole A, Rajeshwari M, Varalakshmi P, Houlden H, Ashokkumar B. Riboflavin Responsive Mitochondrial Dysfunction in Neurodegenerative Diseases. J Clin Med. 2017;6:52.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in RCA: 70]  [Article Influence: 8.8]  [Reference Citation Analysis (1)]
50.  Mosegaard S, Dipace G, Bross P, Carlsen J, Gregersen N, Olsen RKJ. Riboflavin Deficiency-Implications for General Human Health and Inborn Errors of Metabolism. Int J Mol Sci. 2020;21:3847.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in RCA: 93]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
51.  Parra M, Stahl S, Hellmann H. Vitamin B₆ and Its Role in Cell Metabolism and Physiology. Cells. 2018;7:84.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in RCA: 227]  [Article Influence: 32.4]  [Reference Citation Analysis (0)]
52.  Sousou JM, Griffith EM, Marsalisi C, Reddy P. Pyridoxine Deficiency and Neurologic Dysfunction: An Unlikely Association. Cureus. 2023;15:e47647.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
53.  Chiabrando D, Fiorito V, Petrillo S, Tolosano E. Unraveling the Role of Heme in Neurodegeneration. Front Neurosci. 2018;12:712.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
54.  Jung HY, Kim W, Hahn KR, Kang MS, Kim TH, Kwon HJ, Nam SM, Chung JY, Choi JH, Yoon YS, Kim DW, Yoo DY, Hwang IK. Pyridoxine Deficiency Exacerbates Neuronal Damage after Ischemia by Increasing Oxidative Stress and Reduces Proliferating Cells and Neuroblasts in the Gerbil Hippocampus. Int J Mol Sci. 2020;21:5551.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
55.  Stockler S, Plecko B, Gospe SM Jr, Coulter-Mackie M, Connolly M, van Karnebeek C, Mercimek-Mahmutoglu S, Hartmann H, Scharer G, Struijs E, Tein I, Jakobs C, Clayton P, Van Hove JL. Pyridoxine dependent epilepsy and antiquitin deficiency: clinical and molecular characteristics and recommendations for diagnosis, treatment and follow-up. Mol Genet Metab. 2011;104:48-60.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in RCA: 202]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
56.  Kaminiów K, Pająk M, Pająk R, Paprocka J. Pyridoxine-Dependent Epilepsy and Antiquitin Deficiency Resulting in Neonatal-Onset Refractory Seizures. Brain Sci. 2021;12:65.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
57.  Wang S, Sun J, Tu Y, Zhu L, Feng Z. Clinical and genetic characteristics of pyridoxine-dependent epilepsy: Case series report of three Chinese patients with phenotypic variability. Exp Ther Med. 2017;14:1989-1992.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
58.  Quadros EV. Folate and Other B Vitamins in Brain Health and Disease. Nutrients. 2023;15:2525.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
59.  Nemati H, Boroujeni AF, Inaloo S, Katibeh P, Shahriari M. The Cerebrospinal Fluid Concentration of Methyltetrahydrofolate and Serum Folate in Children with Developmental Delay, Regression, and/or Refractory Epilepsy. Neurol India. 2021;69:1343-1348.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
60.  Morrell MJ. Folic Acid and Epilepsy. Epilepsy Curr. 2002;2:31-34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
61.  Reynolds EH. The neurology of folic acid deficiency. Handb Clin Neurol. 2014;120:927-943.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in RCA: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
62.  Hirono H, Wada Y. Effects of dietary folate deficiency on developmental increase of myelin lipids in rat brain. J Nutr. 1978;108:766-772.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 16]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
63.  Zhou Y, Cong Y, Liu H. Folic acid ameliorates depression-like behaviour in a rat model of chronic unpredictable mild stress. BMC Neurosci. 2020;21:1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
64.  Greenberg JA, Bell SJ, Guan Y, Yu YH. Folic Acid supplementation and pregnancy: more than just neural tube defect prevention. Rev Obstet Gynecol. 2011;4:52-59.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Briani C, Dalla Torre C, Citton V, Manara R, Pompanin S, Binotto G, Adami F. Cobalamin deficiency: clinical picture and radiological findings. Nutrients. 2013;5:4521-4539.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 156]  [Cited by in RCA: 169]  [Article Influence: 14.1]  [Reference Citation Analysis (0)]
66.  Charters PFP, Morrison HD, Witherick J, King S. Subacute combined degeneration of the cord secondary to nitrous oxide misuse: No laughing matter. BJR Case Rep. 2021;7:20200179.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
67.  Obeid R, Herrmann W. Mechanisms of homocysteine neurotoxicity in neurodegenerative diseases with special reference to dementia. FEBS Lett. 2006;580:2994-3005.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 333]  [Cited by in RCA: 353]  [Article Influence: 18.6]  [Reference Citation Analysis (0)]
68.  Lominadze D, Tyagi N, Sen U, Ovechkin A, Tyagi SC. Homocysteine alters cerebral microvascular integrity and causes remodeling by antagonizing GABA-A receptor. Mol Cell Biochem. 2012;371:89-96.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 21]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
69.  Pushpakumar S, Kundu S, Sen U. Endothelial dysfunction: the link between homocysteine and hydrogen sulfide. Curr Med Chem. 2014;21:3662-3672.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in RCA: 142]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
70.  Azzini E, Raguzzini A, Polito A. A Brief Review on Vitamin B(12) Deficiency Looking at Some Case Study Reports in Adults. Int J Mol Sci. 2021;22:9694.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
71.  Dalangin R, Kim A, Campbell RE. The Role of Amino Acids in Neurotransmission and Fluorescent Tools for Their Detection. Int J Mol Sci. 2020;21:6197.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in RCA: 90]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
72.  Black MM. Effects of vitamin B12 and folate deficiency on brain development in children. Food Nutr Bull. 2008;29:S126-S131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 238]  [Cited by in RCA: 257]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
73.  Kirik S, Çatak Z. Vitamin B12 Deficiency Observed in Children With First Afebrile Seizures. Cureus. 2021;13:e13745.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
74.  Isik U, Beyatli S. Epileptic spasms and partial seizures associated with vitamin B12 deficiency: case report and literature review. Epilepsi. 2017;23:77-80.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
75.  Portillo S, Guevara NA, Francis-Morel G. Psychosis and Seizures Attributed to Severe Vitamin B12 Deficiency: A Case Report. Cureus. 2023;15:e39889.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
76.  Sawicka-Glazer E, Czuczwar SJ. Vitamin C: a new auxiliary treatment of epilepsy? Pharmacol Rep. 2014;66:529-533.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
77.  Ballaz SJ, Rebec GV. Neurobiology of vitamin C: Expanding the focus from antioxidant to endogenous neuromodulator. Pharmacol Res. 2019;146:104321.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 55]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
78.  Santos IM, Tomé Ada R, Saldanha GB, Ferreira PM, Militão GC, Freitas RM. Oxidative stress in the hippocampus during experimental seizures can be ameliorated with the antioxidant ascorbic acid. Oxid Med Cell Longev. 2009;2:214-221.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in RCA: 82]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
79.  Ullah I, Badshah H, Naseer MI, Lee HY, Kim MO. Thymoquinone and vitamin C attenuates pentylenetetrazole-induced seizures via activation of GABAB1 receptor in adult rats cortex and hippocampus. Neuromolecular Med. 2015;17:35-46.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
80.  Domith I, Socodato R, Portugal CC, Munis AF, Duarte-Silva AT, Paes-de-Carvalho R. Vitamin C modulates glutamate transport and NMDA receptor function in the retina. J Neurochem. 2018;144:408-420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
81.  Das A, Sarwar MS, Hossain MS, Karmakar P, Islam MS, Hussain ME, Banik S. Elevated Serum Lipid Peroxidation and Reduced Vitamin C and Trace Element Concentrations Are Correlated With Epilepsy. Clin EEG Neurosci. 2019;50:63-72.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
82.  Bonnet U. The sour side of vitamin C might mediate neuroprotective, anticonvulsive and antidepressant-like effects. Med Hypotheses. 2019;131:109320.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
83.  Shin EJ, Jeong JH, Chung YH, Kim WK, Ko KH, Bach JH, Hong JS, Yoneda Y, Kim HC. Role of oxidative stress in epileptic seizures. Neurochem Int. 2011;59:122-137.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 305]  [Cited by in RCA: 301]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
84.  Nazar AK hamid. The effect of antioxidant supplementation in the treatment of epilepsy. Iraqi J Pharm. 2011;11:27-33.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
85.  Zeng X, Dong X, Xiao Q, Yao J. Vitamin C Inhibits Ubiquitination of Glutamate Transporter 1 (GLT-1) in Astrocytes by Downregulating HECTD1. ACS Chem Neurosci. 2022;13:676-687.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
86.  Warner TA, Kang JQ, Kennard JA, Harrison FE. Low brain ascorbic acid increases susceptibility to seizures in mouse models of decreased brain ascorbic acid transport and Alzheimer's disease. Epilepsy Res. 2015;110:20-25.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
87.  Sharma Y, Popescu A, Horwood C, Hakendorf P, Thompson C. Relationship between Vitamin C Deficiency and Cognitive Impairment in Older Hospitalised Patients: A Cross-Sectional Study. Antioxidants (Basel). 2022;11:463.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 9]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
88.  Hii CS, Ferrante A. The Non-Genomic Actions of Vitamin D. Nutrients. 2016;8:135.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in RCA: 188]  [Article Influence: 20.9]  [Reference Citation Analysis (0)]
89.  Khazai N, Judd SE, Tangpricha V. Calcium and vitamin D: skeletal and extraskeletal health. Curr Rheumatol Rep. 2008;10:110-117.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in RCA: 201]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
90.  Wion D, MacGrogan D, Neveu I, Jehan F, Houlgatte R, Brachet P. 1,25-Dihydroxyvitamin D3 is a potent inducer of nerve growth factor synthesis. J Neurosci Res. 1991;28:110-114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in RCA: 141]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
91.  Wrzosek M, Łukaszkiewicz J, Wrzosek M, Jakubczyk A, Matsumoto H, Piątkiewicz P, Radziwoń-Zaleska M, Wojnar M, Nowicka G. Vitamin D and the central nervous system. Pharmacol Rep. 2013;65:271-278.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in RCA: 118]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
92.  Ko P, Burkert R, McGrath J, Eyles D. Maternal vitamin D3 deprivation and the regulation of apoptosis and cell cycle during rat brain development. Brain Res Dev Brain Res. 2004;153:61-68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in RCA: 100]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
93.  Jiang H, Zhang S. Therapeutic effect of acute and chronic use of different doses of vitamin D3 on seizure responses and cognitive impairments induced by pentylenetetrazole in immature male rats. Iran J Basic Med Sci. 2022;25:84-95.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
94.  Siegel A, Malkowitz L, Moskovits MJ, Christakos S. Administration of 1,25-dihydroxyvitamin D3 results in the elevation of hippocampal seizure threshold levels in rats. Brain Res. 1984;298:125-129.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in RCA: 48]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
95.  Kalueff AV, Minasyan A, Keisala T, Kuuslahti M, Miettinen S, Tuohimaa P. Increased severity of chemically induced seizures in mice with partially deleted Vitamin D receptor gene. Neurosci Lett. 2006;394:69-73.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 47]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
96.  Elmazny A, Amer H, Rashed L, Khalil S, Magdy R. Vitamin D status of untreated children and adolescent Egyptian patients with genetic generalized epilepsy: A case-control study. Epilepsy Behav. 2020;103:106840.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
97.  Holló A, Clemens Z, Kamondi A, Lakatos P, Szűcs A. Correction of vitamin D deficiency improves seizure control in epilepsy: a pilot study. Epilepsy Behav. 2012;24:131-133.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in RCA: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
98.  Alhaidari HM, Babtain F, Alqadi K, Bouges A, Baeesa S, Al-Said YA. Association between serum vitamin D levels and age in patients with epilepsy: a retrospective study from an epilepsy center in Saudi Arabia. Ann Saudi Med. 2022;42:262-268.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
99.  Leandro-Merhi VA, de Almeida Souza Tedrus GM, Jacober de Moraes GG, Ravelli MN. Interaction between vitamin D level, antiseizure medications (ASM) and seizure control in epilepsy adult patients. Rev Neurol (Paris). 2023;179:1111-1117.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
100.  Teagarden DL, Meador KJ, Loring DW. Low vitamin D levels are common in patients with epilepsy. Epilepsy Res. 2014;108:1352-1356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in RCA: 56]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
101.  Wagner CL, Hollis BW. The Implications of Vitamin D Status During Pregnancy on Mother and her Developing Child. Front Endocrinol (Lausanne). 2018;9:500.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in RCA: 82]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
102.  Specht IO, Thorsteinsdottir F, Walker KC, Olsen J, Heitmann BL. Neonatal vitamin D status and risk of childhood epilepsy. Epilepsia. 2020;61:1282-1290.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
103.  Junges C, Machado TD, Nunes Filho PRS, Riesgo R, Mello ED. Vitamin D deficiency in pediatric patients using antiepileptic drugs: systematic review with meta-analysis. J Pediatr (Rio J). 2020;96:559-568.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
104.  Mardones P, Rigotti A. Cellular mechanisms of vitamin E uptake: relevance in alpha-tocopherol metabolism and potential implications for disease. J Nutr Biochem. 2004;15:252-260.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in RCA: 74]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
105.  Mehvari J, Motlagh FG, Najafi M, Ghazvini MR, Naeini AA, Zare M. Effects of Vitamin E on seizure frequency, electroencephalogram findings, and oxidative stress status of refractory epileptic patients. Adv Biomed Res. 2016;5:36.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in RCA: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
106.  Feng J, Zheng Y, Guo M, Ares I, Martínez M, Lopez-Torres B, Martínez-Larrañaga MR, Wang X, Anadón A, Martínez MA. Oxidative stress, the blood-brain barrier and neurodegenerative diseases: The critical beneficial role of dietary antioxidants. Acta Pharm Sin B. 2023;13:3988-4024.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 42]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
107.  Lee KH, Cha M, Lee BH. Neuroprotective Effect of Antioxidants in the Brain. Int J Mol Sci. 2020;21:7152.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in RCA: 246]  [Article Influence: 49.2]  [Reference Citation Analysis (0)]
108.  Ambrogini P, Minelli A, Galati C, Betti M, Lattanzi D, Ciffolilli S, Piroddi M, Galli F, Cuppini R. Post-seizure α-tocopherol treatment decreases neuroinflammation and neuronal degeneration induced by status epilepticus in rat hippocampus. Mol Neurobiol. 2014;50:246-256.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in RCA: 41]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
109.  Betti M, Minelli A, Ambrogini P, Ciuffoli S, Viola V, Galli F, Canonico B, Lattanzi D, Colombo E, Sestili P, Cuppini R. Dietary supplementation with α-tocopherol reduces neuroinflammation and neuronal degeneration in the rat brain after kainic acid-induced status epilepticus. Free Radic Res. 2011;45:1136-1142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in RCA: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
110.  Tomé AR, Feng D, Freitas RM. The effects of alpha-tocopherol on hippocampal oxidative stress prior to in pilocarpine-induced seizures. Neurochem Res. 2010;35:580-587.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in RCA: 51]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
111.  Gabsi S, Gouider-Khouja N, Belal S, Fki M, Kefi M, Turki I, Ben Hamida M, Kayden H, Mebazaa R, Hentati F. Effect of vitamin E supplementation in patients with ataxia with vitamin E deficiency. Eur J Neurol. 2001;8:477-481.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in RCA: 85]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
112.  Di Donato I, Bianchi S, Federico A. Ataxia with vitamin E deficiency: update of molecular diagnosis. Neurol Sci. 2010;31:511-515.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in RCA: 75]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
113.  Copp RP, Wisniewski T, Hentati F, Larnaout A, Ben Hamida M, Kayden HJ. Localization of alpha-tocopherol transfer protein in the brains of patients with ataxia with vitamin E deficiency and other oxidative stress related neurodegenerative disorders. Brain Res. 1999;822:80-87.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in RCA: 99]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
114.  Puri V, Chaudhry N, Tatke M, Prakash V. Isolated vitamin E deficiency with demyelinating neuropathy. Muscle Nerve. 2005;32:230-235.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 21]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
115.  Li J, Lin JC, Wang H, Peterson JW, Furie BC, Furie B, Booth SL, Volpe JJ, Rosenberg PA. Novel role of vitamin k in preventing oxidative injury to developing oligodendrocytes and neurons. J Neurosci. 2003;23:5816-5826.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in RCA: 159]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
116.  Ferland G. Vitamin K and the nervous system: an overview of its actions. Adv Nutr. 2012;3:204-212.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in RCA: 122]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
117.  Ferland G. Vitamin K, an emerging nutrient in brain function. Biofactors. 2012;38:151-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
118.  Rahn JJ, Bestman JE, Josey BJ, Inks ES, Stackley KD, Rogers CE, Chou CJ, Chan SS. Novel Vitamin K analogs suppress seizures in zebrafish and mouse models of epilepsy. Neuroscience. 2014;259:142-154.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
119.  Fiorentini D, Cappadone C, Farruggia G, Prata C. Magnesium: Biochemistry, Nutrition, Detection, and Social Impact of Diseases Linked to Its Deficiency. Nutrients. 2021;13:1136.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in RCA: 185]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
120.  Chen S, Xu D, Fan L, Fang Z, Wang X, Li M. Roles of N-Methyl-D-Aspartate Receptors (NMDARs) in Epilepsy. Front Mol Neurosci. 2021;14:797253.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
121.  Yuen AW, Sander JW. Can magnesium supplementation reduce seizures in people with epilepsy? A hypothesis. Epilepsy Res. 2012;100:152-156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
122.  Baek SJ, Byeon JH, Eun SH, Eun BL, Kim GH. Risk of low serum levels of ionized magnesium in children with febrile seizure. BMC Pediatr. 2018;18:297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
123.  Mathew AA, Panonnummal R. 'Magnesium'-the master cation-as a drug-possibilities and evidences. Biometals. 2021;34:955-986.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 37]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
124.  Abdelmalik PA, Politzer N, Carlen PL. Magnesium as an effective adjunct therapy for drug resistant seizures. Can J Neurol Sci. 2012;39:323-327.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
125.  Guo X, Zhu Y, Ying C, Xu K, Hong Y. The impact of serum magnesium and calcium on the risk of epilepsy: A mendelian randomization study. CNS Neurosci Ther. 2023;29:3062-3067.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
126.  Abdullahi I, Watila MM, Shahi N, Nyandaiti YW, Bwala SA. Serum magnesium in adult patients with idiopathic and symptomatic epilepsy in Maiduguri, Northeast Nigeria. Niger J Clin Pract. 2019;22:186-193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 3]  [Reference Citation Analysis (0)]
127.  Osborn KE, Shytle RD, Frontera AT, Soble JR, Schoenberg MR. Addressing potential role of magnesium dyshomeostasis to improve treatment efficacy for epilepsy: A reexamination of the literature. J Clin Pharmacol. 2016;56:260-265.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
128.  Doboszewska U, Młyniec K, Wlaź A, Poleszak E, Nowak G, Wlaź P. Zinc signaling and epilepsy. Pharmacol Ther. 2019;193:156-177.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
129.  Vergnano AM, Rebola N, Savtchenko LP, Pinheiro PS, Casado M, Kieffer BL, Rusakov DA, Mulle C, Paoletti P. Zinc dynamics and action at excitatory synapses. Neuron. 2014;82:1101-1114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in RCA: 156]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
130.  Choi S, Hong DK, Choi BY, Suh SW. Zinc in the Brain: Friend or Foe? Int J Mol Sci. 2020;21:8941.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 60]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
131.  Saghazadeh A, Mahmoudi M, Meysamie A, Gharedaghi M, Zamponi GW, Rezaei N. Possible role of trace elements in epilepsy and febrile seizures: a meta-analysis. Nutr Rev. 2015;73:760-779.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
132.  Kheradmand Z, Yarali B, Zare A, Pourpak Z, Shams S, Ashrafi MR. Comparison of Serum Zinc and Copper levels in Children and adolescents with Intractable and Controlled Epilepsy. Iran J Child Neurol. 2014;8:49-54.  [PubMed]  [DOI]  [Cited in This Article: ]
133.  Saad K, Hammad E, Hassan AF, Badry R. Trace element, oxidant, and antioxidant enzyme values in blood of children with refractory epilepsy. Int J Neurosci. 2014;124:181-186.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 32]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
134.  Chen NN, Zhao DJ, Sun YX, Wang DD, Ni H. Long-Term Effects of Zinc Deficiency and Zinc Supplementation on Developmental Seizure-Induced Brain Damage and the Underlying GPR39/ZnT-3 and MBP Expression in the Hippocampus. Front Neurosci. 2019;13:920.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
135.  Blakemore LJ, Trombley PQ. Zinc as a Neuromodulator in the Central Nervous System with a Focus on the Olfactory Bulb. Front Cell Neurosci. 2017;11:297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in RCA: 56]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
136.  Rajakulendran S, Hanna MG. The Role of Calcium Channels in Epilepsy. Cold Spring Harb Perspect Med. 2016;6:a022723.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in RCA: 71]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
137.  Südhof TC. Calcium control of neurotransmitter release. Cold Spring Harb Perspect Biol. 2012;4:a011353.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in RCA: 327]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
138.  Clarke SG, Scarnati MS, Paradiso KG. Neurotransmitter Release Can Be Stabilized by a Mechanism That Prevents Voltage Changes Near the End of Action Potentials from Affecting Calcium Currents. J Neurosci. 2016;36:11559-11572.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
139.  Xu JH, Tang FR. Voltage-Dependent Calcium Channels, Calcium Binding Proteins, and Their Interaction in the Pathological Process of Epilepsy. Int J Mol Sci. 2018;19:2735.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 42]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
140.  Stampanoni Bassi M, Iezzi E, Gilio L, Centonze D, Buttari F. Synaptic Plasticity Shapes Brain Connectivity: Implications for Network Topology. Int J Mol Sci. 2019;20:6193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in RCA: 93]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
141.  Han P, Trinidad BJ, Shi J. Hypocalcemia-induced seizure: demystifying the calcium paradox. ASN Neuro. 2015;7:1759091415578050.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in RCA: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
142.  Delorenzo RJ, Sun DA, Deshpande LS. Cellular mechanisms underlying acquired epilepsy: the calcium hypothesis of the induction and maintainance of epilepsy. Pharmacol Ther. 2005;105:229-266.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in RCA: 203]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
143.  George AL Jr. Inherited Channelopathies Associated with Epilepsy. Epilepsy Curr. 2004;4:65-70.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in RCA: 63]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
144.  Wang J, Ou SW, Wang YJ. Distribution and function of voltage-gated sodium channels in the nervous system. Channels (Austin). 2017;11:534-554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in RCA: 100]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
145.  Kaplan DI, Isom LL, Petrou S. Role of Sodium Channels in Epilepsy. Cold Spring Harb Perspect Med. 2016;6:a022814.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in RCA: 74]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
146.  Ragsdale DS, Avoli M. Sodium channels as molecular targets for antiepileptic drugs. Brain Res Brain Res Rev. 1998;26:16-28.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in RCA: 155]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
147.  Nardone R, Brigo F, Trinka E. Acute Symptomatic Seizures Caused by Electrolyte Disturbances. J Clin Neurol. 2016;12:21-33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 107]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
148.  Humphries ES, Dart C. Neuronal and Cardiovascular Potassium Channels as Therapeutic Drug Targets: Promise and Pitfalls. J Biomol Screen. 2015;20:1055-1073.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in RCA: 53]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
149.  Kim DM, Nimigean CM. Voltage-Gated Potassium Channels: A Structural Examination of Selectivity and Gating. Cold Spring Harb Perspect Biol. 2016;8:a029231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in RCA: 92]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
150.  Villa C, Combi R. Potassium Channels and Human Epileptic Phenotypes: An Updated Overview. Front Cell Neurosci. 2016;10:81.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in RCA: 95]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
151.  Köhling R, Wolfart J. Potassium Channels in Epilepsy. Cold Spring Harb Perspect Med. 2016;6:a022871.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in RCA: 99]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
152.  de Curtis M, Uva L, Gnatkovsky V, Librizzi L. Potassium dynamics and seizures: Why is potassium ictogenic? Epilepsy Res. 2018;143:50-59.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
153.  Kaul N, Nicolo J, O’brien TJ, Kwan P. Practical considerations of dietary therapies for epilepsy in adults. Acta Epileptologica. 2021;3:16.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
154.  Abbaspour N, Hurrell R, Kelishadi R. Review on iron and its importance for human health. J Res Med Sci. 2014;19:164-174.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Hare D, Ayton S, Bush A, Lei P. A delicate balance: Iron metabolism and diseases of the brain. Front Aging Neurosci. 2013;5:34.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 288]  [Cited by in RCA: 299]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
156.  Ferreira A, Neves P, Gozzelino R. Multilevel Impacts of Iron in the Brain: The Cross Talk between Neurophysiological Mechanisms, Cognition, and Social Behavior. Pharmaceuticals (Basel). 2019;12:126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 64]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
157.  Habibian N, Alipour A, Rezaianzadeh A. Association between Iron Deficiency Anemia and Febrile Convulsion in 3- to 60-Month-Old Children: A Systematic Review and Meta-Analysis. Iran J Med Sci. 2014;39:496-505.  [PubMed]  [DOI]  [Cited in This Article: ]
158.  Sharif MR, Kheirkhah D, Madani M, Kashani HH. The Relationship Between Iron Deficiency and Febrile Convulsion: A Case-Control Study. Glob J Health Sci. 2015;8:185-189.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
159.  Bidabadi E, Mashouf M. Association between iron deficiency anemia and first febrile convulsion: A case-control study. Seizure. 2009;18:347-351.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in RCA: 25]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
160.  Zimmer TS, David B, Broekaart DWM, Schidlowski M, Ruffolo G, Korotkov A, van der Wel NN, van Rijen PC, Mühlebner A, van Hecke W, Baayen JC, Idema S, François L, van Eyll J, Dedeurwaerdere S, Kessels HW, Surges R, Rüber T, Gorter JA, Mills JD, van Vliet EA, Aronica E. Seizure-mediated iron accumulation and dysregulated iron metabolism after status epilepticus and in temporal lobe epilepsy. Acta Neuropathol. 2021;142:729-759.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 36]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
161.  Chen S, Chen Y, Zhang Y, Kuang X, Liu Y, Guo M, Ma L, Zhang D, Li Q. Iron Metabolism and Ferroptosis in Epilepsy. Front Neurosci. 2020;14:601193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in RCA: 67]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
162.  Huang Z, Rose AH, Hoffmann PR. The role of selenium in inflammation and immunity: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal. 2012;16:705-743.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 484]  [Cited by in RCA: 577]  [Article Influence: 44.4]  [Reference Citation Analysis (0)]
163.  Zoidis E, Seremelis I, Kontopoulos N, Danezis GP. Selenium-Dependent Antioxidant Enzymes: Actions and Properties of Selenoproteins. Antioxidants (Basel). 2018;7:66.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in RCA: 237]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
164.  Tinggi U. Selenium: its role as antioxidant in human health. Environ Health Prev Med. 2008;13:102-108.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in RCA: 337]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
165.  Liang X, Xue Z, Zheng Y, Li S, Zhou L, Cao L, Zou Y. Selenium supplementation enhanced the expression of selenoproteins in hippocampus and played a neuroprotective role in LPS-induced neuroinflammation. Int J Biol Macromol. 2023;234:123740.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 14]  [Reference Citation Analysis (0)]
166.  Solovyev ND. Importance of selenium and selenoprotein for brain function: From antioxidant protection to neuronal signalling. J Inorg Biochem. 2015;153:1-12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in RCA: 172]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
167.  Treiman DM. GABAergic mechanisms in epilepsy. Epilepsia. 2001;42 Suppl 3:8-12.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 494]  [Cited by in RCA: 574]  [Article Influence: 23.9]  [Reference Citation Analysis (0)]
168.  Ashrafi MR, Shabanian R, Abbaskhanian A, Nasirian A, Ghofrani M, Mohammadi M, Zamani GR, Kayhanidoost Z, Ebrahimi S, Pourpak Z. Selenium and intractable epilepsy: is there any correlation? Pediatr Neurol. 2007;36:25-29.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 39]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
169.  Wysoczański T, Sokoła-Wysoczańska E, Pękala J, Lochyński S, Czyż K, Bodkowski R, Herbinger G, Patkowska-Sokoła B, Librowski T. Omega-3 Fatty Acids and their Role in Central Nervous System - A Review. Curr Med Chem. 2016;23:816-831.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in RCA: 69]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
170.  Tejada S, Martorell M, Capó X, Tur JA, Pons A, Sureda A. Omega-3 Fatty Acids in the Management of Epilepsy. Curr Top Med Chem. 2016;16:1897-1905.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
171.  El-Ansary AK, Al-Daihan SK, El-Gezeery AR. On the protective effect of omega-3 against propionic acid-induced neurotoxicity in rat pups. Lipids Health Dis. 2011;10:142.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in RCA: 33]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
172.  Pourmasoumi M, Vosoughi N, Derakhshandeh-Rishehri SM, Assarroudi M, Heidari-Beni M. Association of Omega-3 Fatty Acid and Epileptic Seizure in Epileptic Patients: A Systematic Review. Int J Prev Med. 2018;9:36.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
173.  Tanaka K, Farooqui AA, Siddiqi NJ, Alhomida AS, Ong WY. Effects of docosahexaenoic Acid on neurotransmission. Biomol Ther (Seoul). 2012;20:152-157.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
174.  Liang Z, Lou Y, Li Z, Liu S. Causal relationship between human blood omega-3 fatty acids and the risk of epilepsy: A two-sample Mendelian randomization study. Front Neurol. 2023;14:1130439.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
175.  Omrani S, Taheri M, Omrani MD, Arsang-Jang S, Ghafouri-Fard S. The effect of omega-3 fatty acids on clinical and paraclinical features of intractable epileptic patients: a triple blind randomized clinical trial. Clin Transl Med. 2019;8:3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
176.  Sohouli MH, Razmpoosh E, Zarrati M, Jaberzadeh S. The effect of omega-3 fatty acid supplementation on seizure frequency in individuals with epilepsy: a systematic review and meta-analysis. Nutr Neurosci. 2022;25:2421-2430.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
177.  Abu-Ouf NM, Jan MM. The influence of fish oil on neurological development and function. Can J Neurol Sci. 2014;41:13-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
178.  Taha AY, Burnham WM, Auvin S. Polyunsaturated fatty acids and epilepsy. Epilepsia. 2010;51:1348-1358.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in RCA: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
179.  Virmani MA, Cirulli M. The Role of l-Carnitine in Mitochondria, Prevention of Metabolic Inflexibility and Disease Initiation. Int J Mol Sci. 2022;23.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 104]  [Article Influence: 34.7]  [Reference Citation Analysis (0)]
180.  Rho JM, Boison D. The metabolic basis of epilepsy. Nat Rev Neurol. 2022;18:333-347.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 115]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
181.  Augustyniak A, Skrzydlewska E. The influence of L-carnitine suplementation on the antioxidative abilities of serum and the central nervous system of ethanol-induced rats. Metab Brain Dis. 2010;25:381-389.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 24]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
182.  Alhasaniah AH. l-carnitine: Nutrition, pathology, and health benefits. Saudi J Biol Sci. 2023;30:103555.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 33]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
183.  Essawy AE, El-Sayed SA, Tousson E, Abd El-Gawad HS, Alhasani RH, Abd Elkader HAE. Anti-kindling effect of Ginkgo biloba leaf extract and L-carnitine in the pentylenetetrazol model of epilepsy. Environ Sci Pollut Res Int. 2022;29:48573-48587.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
184.  Okumura A, Takagi M, Numoto S, Iwayama H, Azuma Y, Kurahashi H. Effects of l-carnitine supplementation in patients with childhood-onset epilepsy prescribed valproate. Epilepsy Behav. 2021;122:108220.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
185.  Belousova ED. [The decreased level of plasma carnitine in patients with epilepsy]. Zh Nevrol Psikhiatr Im S S Korsakova. 2017;117:106-110.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
186.  De Vivo DC, Bohan TP, Coulter DL, Dreifuss FE, Greenwood RS, Nordli DR Jr, Shields WD, Stafstrom CE, Tein I. L-carnitine supplementation in childhood epilepsy: current perspectives. Epilepsia. 1998;39:1216-1225.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in RCA: 100]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
187.  El Mously S, Abdel Ghaffar H, Magdy R, Hamza S, Mansour M. Carnitine deficiency in epileptic children treated with a diversity of anti-epileptic regimens. Egypt J Neurol Psychiatr Neurosurg. 2018;54:37.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
188.  Bartolini E, Ferrari AR, Fiori S, Della Vecchia S. Glycaemic Imbalances in Seizures and Epilepsy of Paediatric Age: A Literature Review. J Clin Med. 2023;12:2580.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
189.  Schauwecker PE. The effects of glycemic control on seizures and seizure-induced excitotoxic cell death. BMC Neurosci. 2012;13:94.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 30]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
190.  Nadeem MD, Memon S, Qureshi K, Farooq U, Memon UA, Aparna F, Kachhadia MP, Shahzeen F, Ali S, Varrassi G, Kumar L, Kumar S, Kumar S, Khatri M. Seizing the Connection: Exploring the Interplay Between Epilepsy and Glycemic Control in Diabetes Management. Cureus. 2023;15:e45606.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Reference Citation Analysis (0)]
191.  van Campen JS, Jansen FE, Pet MA, Otte WM, Hillegers MH, Joels M, Braun KP. Relation between stress-precipitated seizures and the stress response in childhood epilepsy. Brain. 2015;138:2234-2248.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 31]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
192.  Yang H, Wu J, Guo R, Peng Y, Zheng W, Liu D, Song Z. Glycolysis in energy metabolism during seizures. Neural Regen Res. 2013;8:1316-1326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
193.  Castilla-Guerra L, del Carmen Fernández-Moreno M, López-Chozas JM, Fernández-Bolaños R. Electrolytes disturbances and seizures. Epilepsia. 2006;47:1990-1998.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in RCA: 99]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
194.  Li ZR, Wang CY, Lin WW, Chen YT, Liu XQ, Jiao Z. Handling Delayed or Missed Dose of Antiseizure Medications: A Model-Informed Individual Remedial Dosing. Neurology. 2023;100:e921-e931.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 4]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
195.  Solanki R, Karande A, Ranganathan P. Emerging role of gut microbiota dysbiosis in neuroinflammation and neurodegeneration. Front Neurol. 2023;14:1149618.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 49]  [Reference Citation Analysis (0)]
196.  Wang Y, Zhuo Z, Wang H. Epilepsy, gut microbiota, and circadian rhythm. Front Neurol. 2023;14:1157358.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
197.  Ding M, Lang Y, Shu H, Shao J, Cui L. Microbiota-Gut-Brain Axis and Epilepsy: A Review on Mechanisms and Potential Therapeutics. Front Immunol. 2021;12:742449.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 58]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
198.  Saeed NK, Al-Beltagi M, Bediwy AS, El-Sawaf Y, Toema O. Gut microbiota in various childhood disorders: Implication and indications. World J Gastroenterol. 2022;28:1875-1901.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in CrossRef: 20]  [Cited by in RCA: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (4)]
199.  Satokari R. High Intake of Sugar and the Balance between Pro- and Anti-Inflammatory Gut Bacteria. Nutrients. 2020;12:1348.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
200.  Laudisi F, Stolfi C, Monteleone G. Impact of Food Additives on Gut Homeostasis. Nutrients. 2019;11:2334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 66]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
201.  Konstantinidis T, Tsigalou C, Karvelas A, Stavropoulou E, Voidarou C, Bezirtzoglou E. Effects of Antibiotics upon the Gut Microbiome: A Review of the Literature. Biomedicines. 2020;8:502.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 73]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
202.  Shil A, Chichger H. Artificial Sweeteners Negatively Regulate Pathogenic Characteristics of Two Model Gut Bacteria, E. coli and E. faecalis. Int J Mol Sci. 2021;22:5228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
203.  Lee C, Lee J, Eor JY, Kwak MJ, Huh CS, Kim Y. Effect of Consumption of Animal Products on the Gut Microbiome Composition and Gut Health. Food Sci Anim Resour. 2023;43:723-750.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
204.  Hamerle M, Ghaeni L, Kowski A, Weissinger F, Holtkamp M. Alcohol Use and Alcohol-Related Seizures in Patients With Epilepsy. Front Neurol. 2018;9:401.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
205.  Hillbom M, Pieninkeroinen I, Leone M. Seizures in alcohol-dependent patients: epidemiology, pathophysiology and management. CNS Drugs. 2003;17:1013-1030.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in RCA: 125]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
206.  Samsonsen C, Myklebust H, Strindler T, Bråthen G, Helde G, Brodtkorb E. The seizure precipitating effect of alcohol: A prospective observational cross-over study. Epilepsy Res. 2018;143:82-89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
207.  Bartolomei F, Azulay J-P, Barrie M, Gastaut J-L. Myoclonic alcoholic epilepsy. Epilepsia. 1996;37:406-409.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
208.  Colrain IM, Nicholas CL, Baker FC. Alcohol and the sleeping brain. Handb Clin Neurol. 2014;125:415-431.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in RCA: 94]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
209.  Rojo-Mira J, Pineda-Álvarez M, Zapata-Ospina JP. Efficacy and Safety of Anticonvulsants for the Inpatient Treatment of Alcohol Withdrawal Syndrome: A Systematic Review and Meta-analysis. Alcohol Alcohol. 2022;57:155-164.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
210.  Rogawski MA. Update on the neurobiology of alcohol withdrawal seizures. Epilepsy Curr. 2005;5:225-230.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in RCA: 50]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
211.  Pelliccia A, Lucarelli S, Frediani T, D'Ambrini G, Cerminara C, Barbato M, Vagnucci B, Cardi E. Partial cryptogenetic epilepsy and food allergy/intolerance. A causal or a chance relationship? Reflections on three clinical cases. Minerva Pediatr. 1999;51:153-157.  [PubMed]  [DOI]  [Cited in This Article: ]
212.  Silverberg JI, Joks R, Durkin HG. Allergic disease is associated with epilepsy in childhood: a US population-based study. Allergy. 2014;69:95-103.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 34]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
213.  He CH, Zhao J, Zhu TT. Association between allergic diseases and epilepsy: A systematic review and meta-analysis. Epilepsy Behav. 2021;116:107770.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
214.  Crayton JW, Stone T, Stein G. Epilepsy precipitated by food sensitivity: report of a case with double-blind placebo-controlled assessment. Clin Electroencephalogr. 1981;12:192-198.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 14]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
215.  Frediani T, Lucarelli S, Pelliccia A, Vagnucci B, Cerminara C, Barbato M, Cardi E. Allergy and childhood epilepsy: a close relationship? Acta Neurol Scand. 2001;104:349-352.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 18]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
216.  Yu W, Freeland DMH, Nadeau KC. Food allergy: immune mechanisms, diagnosis and immunotherapy. Nat Rev Immunol. 2016;16:751-765.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 383]  [Cited by in RCA: 391]  [Article Influence: 43.4]  [Reference Citation Analysis (0)]
217.  Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011;7:31-40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1085]  [Cited by in RCA: 1346]  [Article Influence: 89.7]  [Reference Citation Analysis (0)]
218.  Theoharides TC, Zhang B. Neuro-inflammation, blood-brain barrier, seizures and autism. J Neuroinflammation. 2011;8:168.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in RCA: 72]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
219.  Briguglio M, Dell'Osso B, Panzica G, Malgaroli A, Banfi G, Zanaboni Dina C, Galentino R, Porta M. Dietary Neurotransmitters: A Narrative Review on Current Knowledge. Nutrients. 2018;10:591.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in RCA: 116]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
220.  Gorjipour H, Darougar S, Mansouri M, Karimzadeh P, Amouzadeh MH, Sohrabi MR. Hypoallergenic diet may control refractory epilepsy in allergic children: A quasi experimental study. Sci Rep. 2019;9:6875.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
221.  Asadi-Pooya AA, Hossein-Zade A. What do nurses and physicians think about the need for specific dietary restrictions in patients with epilepsy? Epilepsy Behav. 2005;6:604-606.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 9]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
222.  Sarlo GL, Kao A, Holton KF. Investigation of the low glutamate diet as an adjunct treatment for pediatric epilepsy: A pilot randomized controlled trial. Seizure. 2023;106:138-147.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
223.  Verrotti A, Iapadre G, Di Francesco L, Zagaroli L, Farello G. Diet in the Treatment of Epilepsy: What We Know So Far. Nutrients. 2020;12:2645.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
224.  Kaufman KR, Sachdeo RC. Caffeinated beverages and decreased seizure control. Seizure. 2003;12:519-521.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 46]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
225.  Chrościńska-Krawczyk M, Jargiełło-Baszak M, Wałek M, Tylus B, Czuczwar SJ. Caffeine and the anticonvulsant potency of antiepileptic drugs: experimental and clinical data. Pharmacol Rep. 2011;63:12-18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in RCA: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
226.  van Koert RR, Bauer PR, Schuitema I, Sander JW, Visser GH. Caffeine and seizures: A systematic review and quantitative analysis. Epilepsy Behav. 2018;80:37-47.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 44]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
227.  Brundege JM, Dunwiddie TV. Metabolic regulation of endogenous adenosine release from single neurons. Neuroreport. 1998;9:3007-3011.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in RCA: 39]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
228.  Gasior M, Borowicz K, Kleinrok Z, Czuczwar SJ. Chronic caffeine and the anticonvulsant potency of antiepileptic drugs against maximal electroshock. Pharmacol Biochem Behav. 1996;54:639-644.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in RCA: 21]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
229.  Luszczki JJ, Zuchora M, Sawicka KM, Kozińska J, Czuczwar SJ. Acute exposure to caffeine decreases the anticonvulsant action of ethosuximide, but not that of clonazepam, phenobarbital and valproate against pentetrazole-induced seizures in mice. Pharmacol Rep. 2006;58:652-659.  [PubMed]  [DOI]  [Cited in This Article: ]
230.  Bonilha L, Li LM. Heavy coffee drinking and epilepsy. Seizure. 2004;13:284-285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 29]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
231.  Butragueño Laiseca L, Toledo Del Castillo B, Miranda Herrero MC. Energy drinks as a trigger factor for seizures in paediatric patients: A case report. Neurologia (Engl Ed). 2019;34:343-345.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
232.  Nagaraja D, Chand RP. Eating epilepsy. Clin Neurol Neurosurg. 1984;86:95-99.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 35]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
233.  Girges C, Vijiaratnam N, Wirth T, Tjoakarfa C, Idaszak J, Seneviratne U. Seizures triggered by eating - A rare form of reflex epilepsy: A systematic review. Seizure. 2020;83:21-31.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
234.  Tényi D, Janszky J, Jeges S, Schulze-Bonhage A. Food intake precipitates seizures in temporal lobe epilepsy. Sci Rep. 2021;11:16515.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
235.  Vercellino F, Siri L, Brisca G, Scala M, Riva A, Severino M, Striano P. Symptomatic eating epilepsy: two novel pediatric patients and review of literature. Ital J Pediatr. 2021;47:137.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
236.  Vogrig A, Versace S, Gigli GL, Fabris M, Honnorat J, Valente M. Eating reflex epilepsy of presumed autoimmune etiology after SARS-CoV-2 vaccination. J Neurol. 2023;270:3289-3293.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
237.  Singh P, Bansal AR, More A, Bhuyan S, Garg A. Eating Epilepsy in North India: Case Series and Its Management. J Epilepsy Res. 2019;9:152-156.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
238.  Gujjar AR, Jacob PC, Ramanchandiran N, Al-Asmi A. Eating Epilepsy in Oman: A case series and report on the efficacy of temporal lobectomy. Sultan Qaboos Univ Med J. 2013;13:156-161.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
239.  Bertoli S, Cardinali S, Veggiotti P, Trentani C, Testolin G, Tagliabue A. Evaluation of nutritional status in children with refractory epilepsy. Nutr J. 2006;5:14.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in RCA: 43]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
240.  Arya R, Gillespie CW, Cnaan A, Devarajan M, Clark P, Shinnar S, Vinks AA, Mizuno K, Glauser TA; Childhood Absence Epilepsy Study Group. Obesity and overweight as CAE comorbidities and differential drug response modifiers. Neurology. 2016;86:1613-1621.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
241.  Sarangi SC, Kaur N, Tripathi M. Need for pharmacoeconomic consideration of antiepileptic drugs monotherapy treatment in persons with epilepsy. Saudi Pharm J. 2020;28:1228-1237.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
242.  Seligman HK, Laraia BA, Kushel MB. Food insecurity is associated with chronic disease among low-income NHANES participants. J Nutr. 2010;140:304-310.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 962]  [Cited by in RCA: 969]  [Article Influence: 64.6]  [Reference Citation Analysis (0)]
243.  Kolstad E, Bjørk M, Gilhus NE, Alfstad K, Clench-Aas J, Lossius M. Young people with epilepsy have an increased risk of eating disorder and poor quality diet. Epilepsia Open. 2018;3:40-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 7]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
244.  Asadi-Pooya AA, Mintzer S, Sperling MR. Nutritional supplements, foods, and epilepsy: is there a relationship? Epilepsia. 2008;49:1819-1827.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 14]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
245.  Dreischmeier E, Zuloaga A, Kotloski RJ, Karasov AO, Gidal BE. Levetiracetam-associated irritability and potential role of vitamin B6 use in veterans with epilepsy. Epilepsy Behav Rep. 2021;16:100452.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Reference Citation Analysis (0)]
246.  Gauthier A, Simic N, Jones KC, RamachandranNair R. Modified Atkins Diet with slow reduction of carbohydrate. Epilepsy Behav Rep. 2020;13:100353.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 2]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
247.  Ben-Menachem E. Weight issues for people with epilepsy--a review. Epilepsia. 2007;48 Suppl 9:42-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in RCA: 143]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
248.  Farahani HN, Ashthiani AR, Masihi MS. Study on serum zinc and selenium levels in epileptic patients. Neurosciences (Riyadh). 2013;18:138-142.  [PubMed]  [DOI]  [Cited in This Article: ]
249.  Coppola G, Ingrosso D, Operto FF, Signoriello G, Lattanzio F, Barone E, Matera S, Verrotti A. Role of folic acid depletion on homocysteine serum level in children and adolescents with epilepsy and different MTHFR C677T genotypes. Seizure. 2012;21:340-343.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
250.  Tayutivutikul N, Wanleenuwat P, Panapongvasin T, Klajing R, Iwanowski P. Dietary effects on antiseizure drug metabolism and management of epilepsy. Seizure. 2022;102:14-21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
251.  Altman R. New oral anticoagulants: are coagulation units still required? Thromb J. 2014;12:3.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
252.  Kennedy DO. B Vitamins and the Brain: Mechanisms, Dose and Efficacy--A Review. Nutrients. 2016;8:68.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 514]  [Cited by in RCA: 495]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
253.  Hernández-Díaz S, Werler MM, Walker AM, Mitchell AA. Folic acid antagonists during pregnancy and the risk of birth defects. N Engl J Med. 2000;343:1608-1614.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 531]  [Cited by in RCA: 435]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
254.  Peraino C, Ehret CF, Groh KR, Meinert JC, D'Arcy-Gomez G. Phenobarbital effects on weight gain and circadian cycling of food intake and body temperature. Proc Soc Exp Biol Med. 1980;165:473-479.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
255.  Venkatesan N, Davidson MB, Simsolo RB, Kern PA. Phenobarbital treatment enhances insulin-mediated glucose metabolism and improves lipid metabolism in the diabetic rat. Metabolism. 1994;43:348-356.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 18]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
256.  Faridi MM, Aggarwal A. Phenytoin induced vitamin D deficiency presenting as proximal muscle weakness. Indian Pediatr. 2010;47:624-625.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in RCA: 5]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
257.  Howe AM, Lipson AH, Sheffield LJ, Haan EA, Halliday JL, Jenson F, David DJ, Webster WS. Prenatal exposure to phenytoin, facial development, and a possible role for vitamin K. Am J Med Genet. 1995;58:238-244.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in RCA: 40]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
258.  Lewis DP, Van Dyke DC, Willhite LA, Stumbo PJ, Berg MJ. Phenytoin-folic acid interaction. Ann Pharmacother. 1995;29:726-735.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in RCA: 62]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
259.  Xu Y, Zhang N, Xu S, Xu H, Chen S, Xia Z. Effects of phenytoin on serum levels of homocysteine, vitamin B12, folate in patients with epilepsy: A systematic review and meta-analysis (PRISMA-compliant article). Medicine (Baltimore). 2019;98:e14844.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
260.  LoPinto-Khoury C, Brennan L, Mintzer S. Impact of carbamazepine on vitamin D levels: A meta-analysis. Epilepsy Res. 2021;178:106829.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
261.  Sharma TK, Vardey SK, Sitaraman S. Serum Homocysteine, Folate, and Vitamin B12 Levels in Carbamazepine Treated Epileptic Children. Clin Lab. 2016;62:1217-1224.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 5]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
262.  French JA, Gazzola DM. New generation antiepileptic drugs: what do they offer in terms of improved tolerability and safety? Ther Adv Drug Saf. 2011;2:141-158.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 79]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
263.  Tjellesen L, Nilas L, Christiansen C. Does carbamazepine cause disturbances in calcium metabolism in epileptic patients? Acta Neurol Scand. 1983;68:13-19.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 48]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
264.  Kaeley N, Kabi A, Bhatia R, Mohanty A. Carbamazepine-induced hyponatremia - A wakeup call. J Family Med Prim Care. 2019;8:1786-1788.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
265.  Martin CK, Han H, Anton SD, Greenway FL, Smith SR. Effect of valproic acid on body weight, food intake, physical activity and hormones: results of a randomized controlled trial. J Psychopharmacol. 2009;23:814-825.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 34]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
266.  Vázquez M, Fagiolino P, Maldonado C, Olmos I, Ibarra M, Alvariza S, Guevara N, Magallanes L, Olano I. Hyperammonemia associated with valproic acid concentrations. Biomed Res Int. 2014;2014:217269.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in RCA: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
267.  Rakitin A. Does Valproic Acid Have Potential in the Treatment of Diabetes Mellitus? Front Endocrinol (Lausanne). 2017;8:147.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
268.  Rakitin A, Kõks S, Haldre S. Valproate modulates glucose metabolism in patients with epilepsy after first exposure. Epilepsia. 2015;56:e172-e175.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
269.  Arora E, Singh H, Gupta YK. Impact of antiepileptic drugs on bone health: Need for monitoring, treatment, and prevention strategies. J Family Med Prim Care. 2016;5:248-253.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 45]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
270.  Linnebank M, Moskau S, Semmler A, Widman G, Stoffel-Wagner B, Weller M, Elger CE. Antiepileptic drugs interact with folate and vitamin B12 serum levels. Ann Neurol. 2011;69:352-359.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in RCA: 84]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
271.  Gelisse P, Juntas-Morales R, Genton P, Hillaire-Buys D, Diaz O, Coubes P, Crespel A. Dramatic weight loss with levetiracetam. Epilepsia. 2008;49:308-315.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 18]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
272.  Shah D, Husain AM. Utility of levetiracetam in patients with subarachnoid hemorrhage. Seizure. 2009;18:676-679.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in RCA: 32]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
273.  Gidal BE, Sheth RD, Magnus L, Herbeuval AF. Levetiracetam does not alter body weight: analysis of randomized, controlled clinical trials. Epilepsy Res. 2003;56:121-126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in RCA: 28]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
274.  Mahdavi A, Naeini AA, Najafi M, Ghazvini M, Maracy M. Vitamin B12 and Folate Status in Patients with Epilepsy Under Levetiracetam Monotherapy. Int J Prev Med. 2019;10:32.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
275.  Meier C, Kraenzlin ME. Antiepileptics and bone health. Ther Adv Musculoskelet Dis. 2011;3:235-243.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
276.  Trunko ME, Schwartz TA, Marzola E, Klein AS, Kaye WH. Lamotrigine use in patients with binge eating and purging, significant affect dysregulation, and poor impulse control. Int J Eat Disord. 2014;47:329-334.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 11]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
277.  Sachs G, Bowden C, Calabrese JR, Ketter T, Thompson T, White R, Bentley B. Effects of lamotrigine and lithium on body weight during maintenance treatment of bipolar I disorder. Bipolar Disord. 2006;8:175-181.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in RCA: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
278.  Costa B, Vale N. Understanding Lamotrigine's Role in the CNS and Possible Future Evolution. Int J Mol Sci. 2023;24:6050.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 5]  [Reference Citation Analysis (0)]
279.  Kanda J, Izumo N, Kobayashi Y, Onodera K, Shimakura T, Yamamoto N, E Takahashi H, Wakabayashi H. Effects of the antiepileptic drugs topiramate and lamotrigine on bone metabolism in rats. Biomed Res. 2017;38:297-305.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 10]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
280.  Moradi S, Kerman SR, Mollabashi M. The effect of topiramate on weight loss in patients with type 2 diabetes. J Res Med Sci. 2013;18:297-302.  [PubMed]  [DOI]  [Cited in This Article: ]
281.  Belotti EA, Taddeo I, Ragazzi M, Pifferini R, Simonetti GD, Bianchetti MG, Ramelli GP. Chronic impact of topiramate on acid-base balance and potassium in childhood. Eur J Paediatr Neurol. 2010;14:445-448.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 11]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
282.  Mirza N, Marson AG, Pirmohamed M. Effect of topiramate on acid-base balance: extent, mechanism and effects. Br J Clin Pharmacol. 2009;68:655-661.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in RCA: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
283.  Heo K, Rhee Y, Lee HW, Lee SA, Shin DJ, Kim WJ, Song HK, Song K, Lee BI. The effect of topiramate monotherapy on bone mineral density and markers of bone and mineral metabolism in premenopausal women with epilepsy. Epilepsia. 2011;52:1884-1889.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in RCA: 50]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
284.  Salek T, Andel I, Kurfurstova I. Topiramate induced metabolic acidosis and kidney stones - a case study. Biochem Med (Zagreb). 2017;27:404-410.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
285.  Shen AL, Lin HL, Tseng YF, Lin HC, Hsu CY, Chou CY. Topiramate may not increase risk of urolithiasis: A nationwide population-based cohort study. Seizure. 2015;29:86-89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
286.  Mintzer S, Skidmore CT, Sperling MR. B-vitamin deficiency in patients treated with antiepileptic drugs. Epilepsy Behav. 2012;24:341-344.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 18]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
287.  Gidal BE, Maly MM, Kowalski JW, Rutecki PA, Pitterle ME, Cook DE. Gabapentin absorption: effect of mixing with foods of varying macronutrient composition. Ann Pharmacother. 1998;32:405-409.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 19]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
288.  Fantinati M, Trnka J, Signor A, Dumond S, Jourdan G, Verwaerde P, Priymenko N. Appetite-stimulating effect of gabapentin vs mirtazapine in healthy cats post-ovariectomy. J Feline Med Surg. 2020;22:1176-1183.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
289.  Finegan A, Mabrouk U, Nelson LA. Gabapentin-induced bilateral lower extremity edema in a patient with pervasive developmental disorder and schizoaffective disorder. Ment Health Clin. 2020;10:250-253.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
290.  PT670. Antiinflammatory and gastric side effect of gabapentin. Int J Neuropsychopharmacology. 2016;19:44-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
291.  Quintero GC. Review about gabapentin misuse, interactions, contraindications and side effects. J Exp Pharmacol. 2017;9:13-21.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in RCA: 69]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
292.  Verma V, Singh N, Singh Jaggi A. Pregabalin in neuropathic pain: evidences and possible mechanisms. Curr Neuropharmacol. 2014;12:44-56.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in RCA: 107]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
293.  Hoppe C, Rademacher M, Hoffmann JM, Schmidt D, Elger CE. Bodyweight gain under pregabalin therapy in epilepsy: mitigation by counseling patients? Seizure. 2008;17:327-332.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
294.  Gallagher R, Apostle N. Peripheral edema with pregabalin. CMAJ. 2013;185:E506.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 17]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
295.  Stein DJ, Bruce Lydiard R, Herman BK, Mandel FS. Impact of gastrointestinal symptoms on response to pregabalin in generalized anxiety disorder: results of a six-study combined analysis. Int Clin Psychopharmacol. 2009;24:126-132.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
296.  Akin B, Kelle B, Kozanoglu E. The Effect of Pregabalin on Bone Metabolism. J Clin Densitom. 2022;25:223-229.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
297.  Sondhi V, Agarwala A, Pandey RM, Chakrabarty B, Jauhari P, Lodha R, Toteja GS, Sharma S, Paul VK, Kossoff E, Gulati S. Efficacy of Ketogenic Diet, Modified Atkins Diet, and Low Glycemic Index Therapy Diet Among Children With Drug-Resistant Epilepsy: A Randomized Clinical Trial. JAMA Pediatr. 2020;174:944-951.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 90]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
298.  Dighriri IM, Alsubaie AM, Hakami FM, Hamithi DM, Alshekh MM, Khobrani FA, Dalak FE, Hakami AA, Alsueaadi EH, Alsaawi LS, Alshammari SF, Alqahtani AS, Alawi IA, Aljuaid AA, Tawhari MQ. Effects of Omega-3 Polyunsaturated Fatty Acids on Brain Functions: A Systematic Review. Cureus. 2022;14:e30091.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
299.  Lampe JW, Navarro SL, Hullar MA, Shojaie A. Inter-individual differences in response to dietary intervention: integrating omics platforms towards personalised dietary recommendations. Proc Nutr Soc. 2013;72:207-218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in RCA: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
300.  Imdad K, Abualait T, Kanwal A, AlGhannam ZT, Bashir S, Farrukh A, Khattak SH, Albaradie R, Bashir S. The Metabolic Role of Ketogenic Diets in Treating Epilepsy. Nutrients. 2022;14:5074.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in RCA: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
301.  Dowis K, Banga S. The Potential Health Benefits of the Ketogenic Diet: A Narrative Review. Nutrients. 2021;13.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in RCA: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
302.  Lutas A, Yellen G. The ketogenic diet: metabolic influences on brain excitability and epilepsy. Trends Neurosci. 2013;36:32-40.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in RCA: 234]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
303.  Pinto A, Bonucci A, Maggi E, Corsi M, Businaro R. Anti-Oxidant and Anti-Inflammatory Activity of Ketogenic Diet: New Perspectives for Neuroprotection in Alzheimer's Disease. Antioxidants (Basel). 2018;7:63.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in RCA: 147]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
304.  Fan Y, Wang H, Liu X, Zhang J, Liu G. Crosstalk between the Ketogenic Diet and Epilepsy: From the Perspective of Gut Microbiota. Mediators Inflamm. 2019;2019:8373060.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 46]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
305.  Martin-McGill KJ, Bresnahan R, Levy RG, Cooper PN. Ketogenic diets for drug-resistant epilepsy. Cochrane Database Syst Rev. 2020;6:CD001903.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 85]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
306.  Gano LB, Patel M, Rho JM. Ketogenic diets, mitochondria, and neurological diseases. J Lipid Res. 2014;55:2211-2228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in RCA: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
307.  Zhu H, Bi D, Zhang Y, Kong C, Du J, Wu X, Wei Q, Qin H. Ketogenic diet for human diseases: the underlying mechanisms and potential for clinical implementations. Signal Transduct Target Ther. 2022;7:11.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in RCA: 133]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
308.  Kossoff EH, Zupec-Kania BA, Amark PE, Ballaban-Gil KR, Christina Bergqvist AG, Blackford R, Buchhalter JR, Caraballo RH, Helen Cross J, Dahlin MG, Donner EJ, Klepper J, Jehle RS, Kim HD, Christiana Liu YM, Nation J, Nordli DR Jr, Pfeifer HH, Rho JM, Stafstrom CE, Thiele EA, Turner Z, Wirrell EC, Wheless JW, Veggiotti P, Vining EP; Charlie Foundation, Practice Committee of the Child Neurology Society;  Practice Committee of the Child Neurology Society;  International Ketogenic Diet Study Group. Optimal clinical management of children receiving the ketogenic diet: recommendations of the International Ketogenic Diet Study Group. Epilepsia. 2009;50:304-317.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in RCA: 335]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
309.  Shaaban S, Al-Beltagi M, El Rashidy O, Nassar M, El Gendy Y. Ketogenic diet in childhood epilepsy: clinical algorithm in a tertiary care center. Front Pediatr. 2023;11:1221781.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
310.  Wells J, Swaminathan A, Paseka J, Hanson C. Efficacy and Safety of a Ketogenic Diet in Children and Adolescents with Refractory Epilepsy-A Review. Nutrients. 2020;12:1809.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in RCA: 83]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
311.  Goswami JN, Sharma S. Current Perspectives On The Role Of The Ketogenic Diet In Epilepsy Management. Neuropsychiatr Dis Treat. 2019;15:3273-3285.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in RCA: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
312.  Batch JT, Lamsal SP, Adkins M, Sultan S, Ramirez MN. Advantages and Disadvantages of the Ketogenic Diet: A Review Article. Cureus. 2020;12:e9639.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
313.  Zarnowska IM. Therapeutic Use of the Ketogenic Diet in Refractory Epilepsy: What We Know and What Still Needs to Be Learned. Nutrients. 2020;12:2616.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
314.  Watanabe M, Tuccinardi D, Ernesti I, Basciani S, Mariani S, Genco A, Manfrini S, Lubrano C, Gnessi L. Scientific evidence underlying contraindications to the ketogenic diet: An update. Obes Rev. 2020;21:e13053.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in RCA: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
315.  Lakshminarayanan K, Agarawal A, Panda PK, Sinha R, Tripathi M, Pandey RM, Gulati S. Efficacy of low glycemic index diet therapy (LGIT) in children aged 2-8 years with drug-resistant epilepsy: A randomized controlled trial. Epilepsy Res. 2021;171:106574.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
316.  Kim TH, Petrou S, Reid CA. Low glycaemic index diet reduces seizure susceptibility in a syndrome-specific mouse model of generalized epilepsy. Epilepsy Res. 2014;108:139-143.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
317.  Karimzadeh P, Sedighi M, Beheshti M, Azargashb E, Ghofrani M, Abdollahe-Gorgi F. Low Glycemic Index Treatment in pediatric refractory epilepsy: the first Middle East report. Seizure. 2014;23:570-572.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
318.  Pfeifer HH, Lyczkowski DA, Thiele EA. Low glycemic index treatment: implementation and new insights into efficacy. Epilepsia. 2008;49 Suppl 8:42-45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in RCA: 57]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
319.  Muzykewicz DA, Lyczkowski DA, Memon N, Conant KD, Pfeifer HH, Thiele EA. Efficacy, safety, and tolerability of the low glycemic index treatment in pediatric epilepsy. Epilepsia. 2009;50:1118-1126.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in RCA: 123]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
320.  Neal E. Ketogenic dietary therapy for epilepsy and other disorders: current perspectives. Nutr Diet Suppl. 2014;6:25-33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
321.  Varjú P, Farkas N, Hegyi P, Garami A, Szabó I, Illés A, Solymár M, Vincze Á, Balaskó M, Pár G, Bajor J, Szűcs Á, Huszár O, Pécsi D, Czimmer J. Low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet improves symptoms in adults suffering from irritable bowel syndrome (IBS) compared to standard IBS diet: A meta-analysis of clinical studies. PLoS One. 2017;12:e0182942.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in RCA: 98]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
322.  Fusco F, Perottoni S, Giordano C, Riva A, Iannone LF, De Caro C, Russo E, Albani D, Striano P. The microbiota-gut-brain axis and epilepsy from a multidisciplinary perspective: Clinical evidence and technological solutions for improvement of in vitro preclinical models. Bioeng Transl Med. 2022;7:e10296.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
323.  Morariu ID, Avasilcai L, Vieriu M, Lupu VV, Morariu BA, Lupu A, Morariu PC, Pop OL, Starcea IM, Trandafir L. Effects of a Low-FODMAP Diet on Irritable Bowel Syndrome in Both Children and Adults-A Narrative Review. Nutrients. 2023;15:2295.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 6]  [Reference Citation Analysis (0)]
324.  Kundu S, Nayak S, Rakshit D, Singh T, Shukla R, Khatri DK, Mishra A. The microbiome-gut-brain axis in epilepsy: pharmacotherapeutic target from bench evidence for potential bedside applications. Eur J Neurol. 2023;30:3557-3567.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 7]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
325.  Muir JG, Gibson PR. The Low FODMAP Diet for Treatment of Irritable Bowel Syndrome and Other Gastrointestinal Disorders. Gastroenterol Hepatol (N Y). 2013;9:450-452.  [PubMed]  [DOI]  [Cited in This Article: ]
326.  Bellini M, Tonarelli S, Nagy AG, Pancetti A, Costa F, Ricchiuti A, de Bortoli N, Mosca M, Marchi S, Rossi A. Low FODMAP Diet: Evidence, Doubts, and Hopes. Nutrients. 2020;12:148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in RCA: 98]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
327.  Löscher W, Potschka H, Sisodiya SM, Vezzani A. Drug Resistance in Epilepsy: Clinical Impact, Potential Mechanisms, and New Innovative Treatment Options. Pharmacol Rev. 2020;72:606-638.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 537]  [Cited by in RCA: 435]  [Article Influence: 87.0]  [Reference Citation Analysis (0)]
328.  Mohammadi M, Meysamie A, Jahanian A. How Do Parents Think about the Effect of Food and Alternative Medicine on their Epileptic Children? Iran J Pediatr. 2010;20:193-198.  [PubMed]  [DOI]  [Cited in This Article: ]
329.  Gargano D, Appanna R, Santonicola A, De Bartolomeis F, Stellato C, Cianferoni A, Casolaro V, Iovino P. Food Allergy and Intolerance: A Narrative Review on Nutritional Concerns. Nutrients. 2021;13:1638.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in RCA: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
330.  Gasmi A, Nasreen A, Menzel A, Gasmi Benahmed A, Pivina L, Noor S, Peana M, Chirumbolo S, Bjørklund G. Neurotransmitters Regulation and Food Intake: The Role of Dietary Sources in Neurotransmission. Molecules. 2022;28:210.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
331.  Sahithi M, Samskruthi M. Nutritional interventions for the prevention and treatment of neurological disorders such as anxiety, bipolar disorder, depression, epilepsy, multiple sclerosis, and schizophrenia. J Neurosci Neurol Disord. 2022;6:052-071.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
332.  Calder PC, Yaqoob P. Omega-3 (n-3) fatty acids, cardiovascular disease and stability of atherosclerotic plaques. Cell Mol Biol (Noisy-le-grand). 2010;56:28-37.  [PubMed]  [DOI]  [Cited in This Article: ]
333.  Schlanger S, Shinitzky M, Yam D. Diet enriched with omega-3 fatty acids alleviates convulsion symptoms in epilepsy patients. Epilepsia. 2002;43:103-104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in RCA: 117]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
334.  Yuen AW, Sander JW, Fluegel D, Patsalos PN, Bell GS, Johnson T, Koepp MJ. Omega-3 fatty acid supplementation in patients with chronic epilepsy: a randomized trial. Epilepsy Behav. 2005;7:253-258.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in RCA: 111]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
335.  DeGiorgio CM, Miller P, Meymandi S, Gornbein JA. n-3 fatty acids (fish oil) for epilepsy, cardiac risk factors, and risk of SUDEP: clues from a pilot, double-blind, exploratory study. Epilepsy Behav. 2008;13:681-684.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 42]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
336.  Scorza FA, Cysneiros RM, Arida RM, Terra VC, Machado HR, Rabello GM, Albuquerque M, Cavalheiro EA. Fish consumption, contaminants and sudden unexpected death in epilepsy: many more benefits than risks. Braz J Biol. 2010;70:665-670.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in RCA: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
337.  Tong Y. Seizures caused by pyridoxine (vitamin B6) deficiency in adults: A case report and literature review. Intractable Rare Dis Res. 2014;3:52-56.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
338.  Coughlin CR, Gospe SM. Pyridoxine‐dependent epilepsy: Current perspectives and questions for future research. Ann Child Neurol Soc. 2023;1:24-37.  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]
339.  Baltrusch S. The Role of Neurotropic B Vitamins in Nerve Regeneration. Biomed Res Int. 2021;2021:9968228.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in RCA: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
340.  Bochyńska A, Lipczyńska-Łojkowska W, Gugała-Iwaniuk M, Lechowicz W, Restel M, Graban A, Lipska B, Ryglewicz D. The effect of vitamin B supplementation on homocysteine metabolism and clinical state of patients with chronic epilepsy treated with carbamazepine and valproic acid. Seizure. 2012;21:276-281.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 16]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
341.  Fakhroo A, Al-Hammadi M, Fakhroo L, Al-Ali F, Snobar R, Al-Beltagi M, Kamal A. The Effect of High-Dose Methyl Vitamin B12 Therapy on Epileptogenesis in Rats: An In Vivo Study. Cureus. 2023;15:e35929.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
342.  O'Leary F, Samman S. Vitamin B12 in health and disease. Nutrients. 2010;2:299-316.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 243]  [Cited by in RCA: 238]  [Article Influence: 15.9]  [Reference Citation Analysis (0)]
343.  Eyles DW, Burne TH, McGrath JJ. Vitamin D, effects on brain development, adult brain function and the links between low levels of vitamin D and neuropsychiatric disease. Front Neuroendocrinol. 2013;34:47-64.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in RCA: 452]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
344.  Wang W, Li Y, Meng X. Vitamin D and neurodegenerative diseases. Heliyon. 2023;9:e12877.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in RCA: 23]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
345.  Martinc B, Grabnar I, Vovk T. Antioxidants as a preventive treatment for epileptic process: a review of the current status. Curr Neuropharmacol. 2014;12:527-550.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in RCA: 77]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
346.  Rizvi S, Raza ST, Ahmed F, Ahmad A, Abbas S, Mahdi F. The role of vitamin e in human health and some diseases. Sultan Qaboos Univ Med J. 2014;14:e157-e165.  [PubMed]  [DOI]  [Cited in This Article: ]
347.  Upaganlawar AB, Wankhede NL, Kale MB, Umare MD, Sehgal A, Singh S, Bhatia S, Al-Harrasi A, Najda A, Nurzyńska-Wierdak R, Bungau S, Behl T. Interweaving epilepsy and neurodegeneration: Vitamin E as a treatment approach. Biomed Pharmacother. 2021;143:112146.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
348.  Ogunmekan AO, Hwang PA. A randomized, double-blind, placebo-controlled, clinical trial of D-alpha-tocopheryl acetate (vitamin E), as add-on therapy, for epilepsy in children. Epilepsia. 1989;30:84-89.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in RCA: 45]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
349.  Raju GB, Behari M, Prasad K, Ahuja GK. Randomized, double-blind, placebo-controlled, clinical trial of D-alpha-tocopherol (vitamin E) as add-on therapy in uncontrolled epilepsy. Epilepsia. 1994;35:368-372.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 22]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
350.  Podszun M, Frank J. Vitamin E-drug interactions: molecular basis and clinical relevance. Nutr Res Rev. 2014;27:215-231.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in RCA: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
351.  Carr AC, Maggini S. Vitamin C and Immune Function. Nutrients. 2017;9:1211.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 662]  [Cited by in RCA: 921]  [Article Influence: 115.1]  [Reference Citation Analysis (0)]
352.  Chambial S, Dwivedi S, Shukla KK, John PJ, Sharma P. Vitamin C in disease prevention and cure: an overview. Indian J Clin Biochem. 2013;28:314-328.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in RCA: 244]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
353.  Harrison FE, May JM. Vitamin C function in the brain: vital role of the ascorbate transporter SVCT2. Free Radic Biol Med. 2009;46:719-730.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in RCA: 424]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
354.  Agus DB, Gambhir SS, Pardridge WM, Spielholz C, Baselga J, Vera JC, Golde DW. Vitamin C crosses the blood-brain barrier in the oxidized form through the glucose transporters. J Clin Invest. 1997;100:2842-2848.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in RCA: 250]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
355.  Longo N, Frigeni M, Pasquali M. Carnitine transport and fatty acid oxidation. Biochim Biophys Acta. 2016;1863:2422-2435.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in RCA: 540]  [Article Influence: 60.0]  [Reference Citation Analysis (0)]
356.  Hussein AM, Adel M, El-Mesery M, Abbas KM, Ali AN, Abulseoud OA. l-Carnitine Modulates Epileptic Seizures in Pentylenetetrazole-Kindled Rats via Suppression of Apoptosis and Autophagy and Upregulation of Hsp70. Brain Sci. 2018;8:45.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
357.  Ferreira GC, McKenna MC. L-Carnitine and Acetyl-L-carnitine Roles and Neuroprotection in Developing Brain. Neurochem Res. 2017;42:1661-1675.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in RCA: 190]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
358.  Campistol J, Chávez B, Vilaseca MA, Artuch R. [Antiepileptic drugs and carnitine]. Rev Neurol. 2000;30 Suppl 1:S105-S109.  [PubMed]  [DOI]  [Cited in This Article: ]
359.  Paquin J, Aouffen M, De Grandpré E, Nadeau R, Langlois D, Mateescu MA. Neuroprotective and cardioprotective actions of an association of pyruvate, vitamin E and fatty acids. Arzneimittelforschung. 2005;55:359-369.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
360.  Schell JC, Olson KA, Jiang L, Hawkins AJ, Van Vranken JG, Xie J, Egnatchik RA, Earl EG, DeBerardinis RJ, Rutter J. A role for the mitochondrial pyruvate carrier as a repressor of the Warburg effect and colon cancer cell growth. Mol Cell. 2014;56:400-413.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in RCA: 278]  [Article Influence: 25.3]  [Reference Citation Analysis (0)]
361.  Desagher S, Glowinski J, Prémont J. Pyruvate protects neurons against hydrogen peroxide-induced toxicity. J Neurosci. 1997;17:9060-9067.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in RCA: 274]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
362.  Fan R, Wang L, Botchway BOA, Zhang Y, Liu X. Protective role of ethyl pyruvate in spinal cord injury by inhibiting the high mobility group box-1/toll-like receptor4/nuclear factor-kappa B signaling pathway. Front Mol Neurosci. 2022;15:1013033.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
363.  Popova I, Malkov A, Ivanov AI, Samokhina E, Buldakova S, Gubkina O, Osypov A, Muhammadiev RS, Zilberter T, Molchanov M, Paskevich S, Zilberter M, Zilberter Y. Metabolic correction by pyruvate halts acquired epilepsy in multiple rodent models. Neurobiol Dis. 2017;106:244-254.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in RCA: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
364.  Simeone KA, Matthews SA, Samson KK, Simeone TA. Targeting deficiencies in mitochondrial respiratory complex I and functional uncoupling exerts anti-seizure effects in a genetic model of temporal lobe epilepsy and in a model of acute temporal lobe seizures. Exp Neurol. 2014;251:84-90.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in RCA: 48]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
365.  Lee SH, Choi BY, Kho AR, Hong DK, Kang BS, Park MK, Lee SH, Choi HC, Song HK, Suh SW. Combined Treatment of Dichloroacetic Acid and Pyruvate Increased Neuronal Survival after Seizure. Nutrients. 2022;14:4804.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 5]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
366.  Sattarinezhad E, Shafaroodi H, Sheikhnouri K, Mousavi Z, Moezi L. The effects of coenzyme Q10 on seizures in mice: the involvement of nitric oxide. Epilepsy Behav. 2014;37:36-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in RCA: 17]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
367.  Sifuentes-Franco S, Sánchez-Macías DC, Carrillo-Ibarra S, Rivera-Valdés JJ, Zuñiga LY, Sánchez-López VA. Antioxidant and Anti-Inflammatory Effects of Coenzyme Q10 Supplementation on Infectious Diseases. Healthcare (Basel). 2022;10:487.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
368.  Bagheri S, Haddadi R, Saki S, Kourosh-Arami M, Rashno M, Mojaver A, Komaki A. Neuroprotective effects of coenzyme Q10 on neurological diseases: a review article. Front Neurosci. 2023;17:1188839.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
369.  Tawfik MK. Coenzyme Q10 enhances the anticonvulsant effect of phenytoin in pilocarpine-induced seizures in rats and ameliorates phenytoin-induced cognitive impairment and oxidative stress. Epilepsy Behav. 2011;22:671-677.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in RCA: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
370.  Mancuso M, Orsucci D, Calsolaro V, Choub A, Siciliano G. Coenzyme Q10 and Neurological Diseases. Pharmaceuticals (Basel). 2009;2:134-149.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
371.  Gupta SC, Patchva S, Aggarwal BB. Therapeutic roles of curcumin: lessons learned from clinical trials. AAPS J. 2013;15:195-218.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1048]  [Cited by in RCA: 1202]  [Article Influence: 92.5]  [Reference Citation Analysis (1)]
372.  Ak T, Gülçin I. Antioxidant and radical scavenging properties of curcumin. Chem Biol Interact. 2008;174:27-37.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1104]  [Cited by in RCA: 1156]  [Article Influence: 68.0]  [Reference Citation Analysis (1)]
373.  Garodia P, Hegde M, Kunnumakkara AB, Aggarwal BB. Curcumin, inflammation, and neurological disorders: How are they linked? Integr Med Res. 2023;12:100968.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 16]  [Reference Citation Analysis (0)]
374.  Dhir A. Curcumin in epilepsy disorders. Phytother Res. 2018;32:1865-1875.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in RCA: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
375.  Choo BKM, Shaikh MF. Mechanism of Curcuma longa and Its Neuroactive Components for the Management of Epileptic Seizures: A Systematic Review. Curr Neuropharmacol. 2021;19:1496-1518.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in RCA: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
376.  Huang C, Chi XS, Li R, Hu X, Xu HX, Li JM, Zhou D. Inhibition of P2X7 Receptor Ameliorates Nuclear Factor-Kappa B Mediated Neuroinflammation Induced by Status Epilepticus in Rat Hippocampus. J Mol Neurosci. 2017;63:173-184.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in RCA: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
377.  Forouzanfar F, Majeed M, Jamialahmadi T, Sahebkar A. Curcumin: A Review of Its Effects on Epilepsy. Adv Exp Med Biol. 2021;1291:363-373.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
378.  Mehla J, Reeta KH, Gupta P, Gupta YK. Protective effect of curcumin against seizures and cognitive impairment in a pentylenetetrazole-kindled epileptic rat model. Life Sci. 2010;87:596-603.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in RCA: 119]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
379.  Agarwal NB, Jain S, Nagpal D, Agarwal NK, Mediratta PK, Sharma KK. Liposomal formulation of curcumin attenuates seizures in different experimental models of epilepsy in mice. Fundam Clin Pharmacol. 2013;27:169-172.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in RCA: 43]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
380.  Jiang Z, Guo M, Shi C, Wang H, Yao L, Liu L, Xie C, Pu S, LaChaud G, Shen J, Zhu M, Mu L, Ge H, Long Y, Wang X, Song Y, Sun J, Hou X, Zarringhalam A, Park SH, Shi C, Shen H, Lin Z. Protection against cognitive impairment and modification of epileptogenesis with curcumin in a post-status epilepticus model of temporal lobe epilepsy. Neuroscience. 2015;310:362-371.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in RCA: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
381.  Hegde M, Girisa S, BharathwajChetty B, Vishwa R, Kunnumakkara AB. Curcumin Formulations for Better Bioavailability: What We Learned from Clinical Trials Thus Far? ACS Omega. 2023;8:10713-10746.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in RCA: 67]  [Article Influence: 33.5]  [Reference Citation Analysis (0)]
382.  Hewlings SJ, Kalman DS. Curcumin: A Review of Its Effects on Human Health. Foods. 2017;6:92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 886]  [Cited by in RCA: 1281]  [Article Influence: 160.1]  [Reference Citation Analysis (0)]
383.  Benameur T, Giacomucci G, Panaro MA, Ruggiero M, Trotta T, Monda V, Pizzolorusso I, Lofrumento DD, Porro C, Messina G. New Promising Therapeutic Avenues of Curcumin in Brain Diseases. Molecules. 2021;27:236.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in RCA: 25]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
384.  Al Alawi AM, Majoni SW, Falhammar H. Magnesium and Human Health: Perspectives and Research Directions. Int J Endocrinol. 2018;2018:9041694.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in RCA: 180]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
385.  Hou H, Wang L, Fu T, Papasergi M, Yule DI, Xia H. Magnesium Acts as a Second Messenger in the Regulation of NMDA Receptor-Mediated CREB Signaling in Neurons. Mol Neurobiol. 2020;57:2539-2550.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
386.  Le Prell CG, Hughes LF, Miller JM. Free radical scavengers vitamins A, C, and E plus magnesium reduce noise trauma. Free Radic Biol Med. 2007;42:1454-1463.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in RCA: 143]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
387.  Yamanaka R, Tabata S, Shindo Y, Hotta K, Suzuki K, Soga T, Oka K. Mitochondrial Mg(2+) homeostasis decides cellular energy metabolism and vulnerability to stress. Sci Rep. 2016;6:30027.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in RCA: 111]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
388.  Herzberg L, Herzeberg B. Mood change and magnesium. A possible interaction between magnesium and lithium? J Nerv Ment Dis. 1977;165:423-426.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in RCA: 11]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
389.  Slutsky I, Abumaria N, Wu LJ, Huang C, Zhang L, Li B, Zhao X, Govindarajan A, Zhao MG, Zhuo M, Tonegawa S, Liu G. Enhancement of learning and memory by elevating brain magnesium. Neuron. 2010;65:165-177.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in RCA: 191]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
390.  Safar MM, Abdallah DM, Arafa NM, Abdel-Aziz MT. Magnesium supplementation enhances the anticonvulsant potential of valproate in pentylenetetrazol-treated rats. Brain Res. 2010;1334:58-64.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in RCA: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
391.  Yadav VK, Amrita A, Yadav S, Kumar R, Yadav KK. Role of Magnesium Supplementation in Children with West Syndrome: A Randomized Controlled Clinical Trial. Iran J Child Neurol. 2022;16:65-75.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
392.  Laskowska M, Bednarek A. Optimizing Delivery Strategies in Eclampsia: A Comprehensive Review on Seizure Management and Birth Methods. Med Sci Monit. 2023;29:e941709.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
393.  Dhande PP, Ranade RS, Ghongane BB. Effect of magnesium oxide on the activity of standard anti-epileptic drugs against experimental seizures in rats. Indian J Pharmacol. 2009;41:268-272.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
394.  Razzaque MS. Magnesium: Are We Consuming Enough? Nutrients. 2018;10.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in RCA: 115]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
395.  Heidt C, Fobker M, Newport M, Feldmann R, Fischer T, Marquardt T. Beta-Hydroxybutyrate (BHB), Glucose, Insulin, Octanoate (C8), and Decanoate (C10) Responses to a Medium-Chain Triglyceride (MCT) Oil with and without Glucose: A Single-Center Study in Healthy Adults. Nutrients. 2023;15:1148.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
396.  Shcherbakova K, Schwarz A, Apryatin S, Karpenko M, Trofimov A. Supplementation of Regular Diet With Medium-Chain Triglycerides for Procognitive Effects: A Narrative Review. Front Nutr. 2022;9:934497.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
397.  Khoramnia A, Ebrahimpour A, Ghanbari R, Ajdari Z, Lai OM. Improvement of medium chain fatty acid content and antimicrobial activity of coconut oil via solid-state fermentation using a Malaysian Geotrichum candidum. Biomed Res Int. 2013;2013:954542.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 19]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
398.  Boateng L, Ansong R, Owusu WB, Steiner-Asiedu M. Coconut oil and palm oil's role in nutrition, health and national development: A review. Ghana Med J. 2016;50:189-196.  [PubMed]  [DOI]  [Cited in This Article: ]
399.  Yu E, Hu FB. Dairy Products, Dairy Fatty Acids, and the Prevention of Cardiometabolic Disease: a Review of Recent Evidence. Curr Atheroscler Rep. 2018;20:24.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in RCA: 76]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
400.  Watanabe S, Tsujino S. Applications of Medium-Chain Triglycerides in Foods. Front Nutr. 2022;9:802805.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in RCA: 10]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
401.  Magdy R, Kishk NA, Abokrysha NT, Ramzy GM, Rizk HI, Hussein M. Fasting and post fasting effect of Ramadan on different seizure types in patients with active epilepsy. Nutr Neurosci. 2022;25:1100-1104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in RCA: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
402.  Vlachos D, Malisova S, Lindberg FA, Karaniki G. Glycemic Index (GI) or Glycemic Load (GL) and Dietary Interventions for Optimizing Postprandial Hyperglycemia in Patients with T2 Diabetes: A Review. Nutrients. 2020;12:1561.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 63]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
403.  Eadie MJ. The risk-benefit ratio of anticonvulsant drugs. Med Toxicol Adverse Drug Exp. 1987;2:324-337.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in RCA: 7]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
404.  Eatock J, Baker GA. Managing patient adherence and quality of life in epilepsy. Neuropsychiatr Dis Treat. 2007;3:117-131.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in RCA: 83]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
405.  Baratta F, Angelico F, Del Ben M. Challenges in Improving Adherence to Diet and Drug Treatment in Hypercholesterolemia Patients. Int J Environ Res Public Health. 2023;20:5878.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
406.  Shyam S, Lee KX, Tan ASW, Khoo TA, Harikrishnan S, Lalani SA, Ramadas A. Effect of Personalized Nutrition on Dietary, Physical Activity, and Health Outcomes: A Systematic Review of Randomized Trials. Nutrients. 2022;14:4104.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in RCA: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
407.  Erickson N, Sullivan ES, Kalliostra M, Laviano A, Wesseling J. Nutrition care is an integral part of patient-centred medical care: a European consensus. Med Oncol. 2023;40:112.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in RCA: 3]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
408.  Adeola OL, Agudosi GM, Akueme NT, Okobi OE, Akinyemi FB, Ononiwu UO, Akunne HS, Akinboro MK, Ogbeifun OE, Okeaya-Inneh M. The Effectiveness of Nutritional Strategies in the Treatment and Management of Obesity: A Systematic Review. Cureus. 2023;15:e45518.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
409.  Bhati D, Deogade MS, Kanyal D. Improving Patient Outcomes Through Effective Hospital Administration: A Comprehensive Review. Cureus. 2023;15:e47731.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 15]  [Reference Citation Analysis (0)]
410.  Islam MR, Trenholm J, Rahman A, Pervin J, Ekström EC, Rahman SM. Sociocultural Influences on Dietary Practices and Physical Activity Behaviors of Rural Adolescents-A Qualitative Exploration. Nutrients. 2019;11:2916.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in RCA: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
411.  Rapacciuolo A, Perrone Filardi P, Cuomo R, Mauriello V, Quarto M, Kisslinger A, Savarese G, Illario M, Tramontano D. The Impact of Social and Cultural Engagement and Dieting on Well-Being and Resilience in a Group of Residents in the Metropolitan Area of Naples. J Aging Res. 2016;2016:4768420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in RCA: 21]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
412.  Johannessen SI, Landmark CJ. Antiepileptic drug interactions - principles and clinical implications. Curr Neuropharmacol. 2010;8:254-267.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in RCA: 220]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
413.  Endris BS, Fenta E, Getnet Y, Spigt M, Dinant GJ, Gebreyesus SH. Barriers and facilitators to the implementation of nutrition interventions at primary health care units of Ethiopia: A consolidated framework for implementation research. Matern Child Nutr. 2023;19:e13433.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
414.  Li T, Steibel JP, Willette AA. Vitamin B6, B12, and Folate's Influence on Neural Networks in the UK Biobank Cohort. Nutrients. 2024;16:2050.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
415.  Ashique S, Mohanto S, Ahmed MG, Mishra N, Garg A, Chellappan DK, Omara T, Iqbal S, Kahwa I. Gut-brain axis: A cutting-edge approach to target neurological disorders and potential synbiotic application. Heliyon. 2024;10:e34092.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited in This Article: ]  [Cited by in RCA: 27]  [Reference Citation Analysis (0)]
416.  Muscaritoli M. The Impact of Nutrients on Mental Health and Well-Being: Insights From the Literature. Front Nutr. 2021;8:656290.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in RCA: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]