ArticlePDF Available

The effects of the ketogenic diet on psychiatric symptomatology, weight and metabolic dysfunction in schizophrenia patients

Authors:
  • Centro de Especialidades Neuropsicológicas Neuroinnova, Ecuador
Research Article
Clinical Nutrition and Metabolism
Clin Nutr Metab, 2018 doi: 10.15761/CNM.1000105 Volume 1(1): 1-5
e eects of the ketogenic diet on psychiatric
symptomatology, weight and metabolic dysfunction in
schizophrenia patients
Javier Gilbert-Jaramillo1*, Dario Vargas-Pico2, Thonny Espinosa-Mendoza2, Svenja Falk2, Kimberly Llanos-Fernández2, Jonathan
Guerrero-Haro1, Carlos Orellana-Román2, Carlos Poveda-Loor1, José Valdevila-Figueira3 and Christopher Palmer4
1ESPOL Polytechnic University, Escuela Superior Politecnica del Litoral, Faculty of Life Sciences, Campus Gustavo Galindo Km 30.5 Via Perimetral, P.O. Box
09-01-5863, Guayaquil, Ecuador, UK
2Institute of Neurosciences of Guayaquil 090514, Ecuador, UK
3Addictive Behavior Unit, Institute of Neurosciences of Guayaquil, UK
4Department of Postgraduate and Continuing Education, McLean hospital Harvard Medical School, USA
Abstract
Objective: e authors aimed to test the eect of a therapeutic ketogenic diet (KD) on the psychotic condition, body composition and metabolic dysfunction in
schizophrenic patients, as well as to report the compliance with the diet.
Method: Two Ecuadorian schizophrenic patients’ male and female (twins) aged 22, were included in a six-week controlled-blinded pilot study under the therapeutic
KD. Compliance was determined by daily urine measurements (commercially available ketone strips). Body composition was analyzed using bio-impedance (Tanita
SC-331S) and the clinical outcomes were assessed by blood, urine and electrocardiogram analysis. e psychiatry condition was evaluated by the PANSS scale and, a
two weeks follow-up after intervention was conducted to evaluate patients’ health condition.
Results: During intervention, after ~15 days of ketosis, PANSS scores decreased in both female (97 to 91) and male (82 to 75) patients. Body fat decreased from
24.5% to 19.8% and 21.7% to 16.8%, respectively. Interestingly, after the third week of the intervention, male patient’ liver enzymes were downregulated to normal
levels (AST=0-40 U/L & ALT=0-41 U/L). No other signicant clinical outcomes were observed during the study. Of relevance, both patients broke the KD in several
occasions.
Conclusions: e present research showed that a short-time ketogenic diet (KD) has positive eects in the psychiatric condition, metabolic dysfunction and body
composition of young schizophrenia patients; suggesting the need of a clinical trial to corroborate its use as a co-treatment.
*Correspondence to: Javier Gilbert-Jaramillo, Department of Physiology,
Anatomy and Genetics; University of Oxford, Oxford OX1 3QX, E-mail:
javier.gilbertjaramillo@dpag.ox.ac.uk
Key words: ketogenic diet, schizophrenia, psychiatric conditions, clinical trial, co-
trearment
Received: July 10, 2018; Accepted: July 25, 2018; Published: August 02, 2018
Introduction
Schizophrenia is a psychiatric disorder with a lifetime prevalence
of ~1 percent and it is characterized by cognitive, positive and negative
aective symptoms [1,2]. Although the etiology of schizophrenia
remains unknown, recent evidence suggests both a hyper-responsive
dopaminergic system in the associative striatum [3] and, mitochondrial
dysfunction and energy metabolism alterations [4,5].
e most commonly prescribed psychopharmacological
intervention for patients with schizophrenia are atypical antipsychotics
(AAP). All AAP currently in use, eectively block dopaminergic D2
receptors, thereby reducing or eliminating the positive symptoms of
schizophrenia [6]. However, they pose serious adverse eects, such
as disturbances of glucose and/or fatty acid metabolism and weight
gain, thus, patients treated with AAPs frequently exhibit increased
co-morbidities of obesity, hyperglycemia, type 2 diabetes mellitus and
dyslipidemia [7-9].
e high fat, low carbohydrate ketogenic diet (KD) was developed
as an eective non-pharmacological treatment for epileptic seizures in
the 1920s [10-13], and more recently is being studied in weight loss
and a variety of neurological and psychiatric disorders [14,15]. e KD
utilizes a high fat (75% of daily intake, DI), extremely low carbohydrates
(lower than 5% of DI) and moderate protein intake (below 20% of
DI) [16,17], to promote the use of fat-derived ketone bodies (KB),
i.e. acetoacetate, –hydroxybutyric acid and acetone, as a non-glucose
source of energy in the brain [18].
Furthermore, a three-week KD conducted in schizophrenia
mice (C57BL/6) not only showed signicant weight loss, but also
demonstrated signicant improvements in the positive, negative
and cognitive symptoms (measured as ataxia, social interaction,
psychomotor hyperactivity, stereotyped behavior, social withdrawal,
and spatial working memory) of the mice [19]. Comparable results
have been described in a recent case report of two patients with
schizoaective disorders, who showed considerable weight loss and
decline in positive and negative symptoms throughout a year-long,
self-prescribed KD [20]. Even though these improvements reversed
when the KD was disrupted, they could be recovered when ketosis was
Gilbert-Jaramillo J (2018) e eects of the ketogenic diet on psychiatric symptomatology, weight and metabolic dysfunction in schizophrenia patients
Volume 1(1): 2-5
Clin Nutr Metab, 2018 doi: 10.15761/CNM.1000105
regained. Similarly, a 1965 pioneer KD trial, examining its eects in
10 treatment-resistant schizophrenic patients, found that symptoms
signicantly decreased aer two weeks following a KD [21]. Although
pioneering, the study neglected to measure ketone levels and drug-
induced adverse eects on metabolic dysfunction could not be
examined, as AAPs had yet to be developed.
us, this underlying pilot study aimed to examine the compliance
and the eects of a controlled six-weeks therapeutic 3:1 ratio KD in
Ecuadorian patients with schizophrenia; a country similar to others
in South American where the typical diet consists of rice, plantain,
potatoes, fruits, pasta, meat, chicken, sh, and vegetable salads; and
carbohydrates constitute about 60% of the DI [22].
Method
Participants
Two 22-year old opposite-sex twins with schizophrenia were
recruited at the outpatient hospital unit of the Institute of Neurosciences
of Guayaquil, Ecuador to participate in a six-week therapeutic 3:1 ratio
ketogenic diet pilot study, from early December 2017. e patients’
parents were still the primary caregivers, responsible for administering
and managing disease treatment and diet.
Study inclusion criteria were set to patients with schizophrenia,
aged between 18 and 30, who have been on their concurrent atypical
antipsychotic intervention for longer than four months. Exclusion
were: pregnancy, lactose intolerance, vegans or vegetarians, patients
with established type 2 diabetes, evidence of cardiovascular disease,
osteoporosis, or kidney/hepatic problems or renal insuciencies.
e female patient was diagnosed with schizophrenia at age 14. Her
medical history showed previous pharmacological intervention with
valproic acid, lorazepam, haloperidol decanoate, levomepromazine,
thioridazine, carbamazepine and uoxetine. Her daily pharmacological
therapy consisted of clozapine (300 mg), risperidone (6 mg),
clonazepam (3 mg) and biperiden (6 mg) for the ve months prior to
the commencement of the study.
e male patient was diagnosed with schizophrenia at age 18 and
his medical history revealed previous pharmacological treatments
with risperidone, biperiden, valproic acid, uoxetine, lorazepam,
clonazepam, lamotrigine and quetiapine. In the 23 months leading
up to the study, his daily pharmacological intervention consisted of
levomepromazine (150 mg), quetiapine (100 mg), valproic acid (1000
mg), biperiden (6 mg) and risperidone (4 mg).
Both patients were maintained on their current pharmacotherapeutic
regimens for the duration of the study
Protocol
Both patients received a 3:1 ratio ketogenic diet plan, which was set
to a daily standard of 2000 kcal and mainly consisted of avocado, olive
oil, butter, eggs, cheese, meat, spinach and broccoli. About 87% of the
daily caloric intake stemmed from fat sources, with the remainder made
up of protein plus carbohydrates [23]. erefore, total net-carbohydrate
intake was calculated to be less than 15 grams per day.
Clinical baseline measurements of body composition,
blood haematological clinical chemistry, urine composition,
electrocardiogram (ECG) and the Positive and Negative Symptom
Scale (PANSS) were examined the day prior to commencing on the
dietary intervention and assessed for ketone bodies (KB; in urine),
serum bilirubin, haematic biometry and minerals (sodium, magnesium
and potassium, which were previously suggested to be aected by KD
intervention) [24-26].
All body composition measurements, blood and urine samples
were obtained at a fasted state at 7.00am, while all ECGs were recorded
at 8 am. Body composition (body fat, muscle mass, bone mass, body
weight and height) was analyzed using a bio-impedance total body
composition analyzer (Tanita SC-331S) and recorded. Both body
composition and ECG examinations were conducted and interpreted by
the same trained professional. PANSS interviews were conducted by the
same trained professional for both patients. Environmental conditions,
i.e. luminosity and noise, were kept constant. e interviewer was
blind to the patients’ compliance to their dietary protocols to avoid any
evaluation bias.
Ketosis was determined through daily measurements of urine
ketone levels at 7 a.m., as per recommendation of Urbain & Bertz [27],
with commercially available ketone strips (Healthy Wiser), as previously
measured ketonuria levels of urine clinical physiochemical analysis
correlated with the commercially available urine strips, conrming its
correct use to determine the nutritional ketotic state [28]. All estimated
KB values were interpreted and recorded by the patients’ parents,
according to the manufacturer’s instructions. Additional clinical urine
analysis of KB was conducted in weekly increments commencing aer
15 days of the start of the study. Blood tests were carried out baseline, on
day 19 and in the last day of the study. Body composition was measured
every 15 days, and the calculated BMI was tracked throughout the
study. ECG was performed at baseline and study conclusion condition.
PANSS interviews took place at baseline and aer day 15 of the study
protocol, in weekly intervals. PANSS score was not recorded during
week four of the protocol due to patient travel. 15 days post-study, when
patients had resumed their regular Ecuadorian diet, blood samples,
body composition and PANSS were measured and analyzed to evaluate
any lasting eects of the KD.
Ethical considerations
e research was approved by the Institute of Neurosciences of
Guayaquil (INC), and National ethics approval was obtained from
the Human Research Ethics Committee of the Hospital Luis Vernaza
(Guayaquil, Ecuador). Both patients and their legal caregivers, i.e.
the parents, were thoroughly briefed on all research objectives, study
conditions, protocols, potential risks and inconveniences and informed
consent was obtained from patients and parents prior to the start of the
study and assent was reconrmed every 15 days before additional blood
and urine samples were obtained.
Results
Diet compliance and Tolerability
Both patients broke the ketogenic diet on several occasions. e
female patient did not fully comply with the prescribed diet for the
rst 21 days of the study and frequently consumed sweets and fruits.
Following non-compliance during the December holiday period, she
fully complied with the ketogenic diet and remained in moderate/high
ketosis for 15 consecutive days. e male patient fully complied with
the diet for the rst three weeks of the study; achieving moderate/high
ketosis within 3 days of commencing the diet. He remained in ketosis
for 18 consecutive days. However, following a beak with the diet during
the December holiday and new year’s evening celebrations, the male
patient was unable to remain in moderate/high ketosis for longer than
~4 consecutive days. Moreover, both patients reported that adherence to
the ketogenic dietary protocol for longer than 14 days was problematic
Gilbert-Jaramillo J (2018) e eects of the ketogenic diet on psychiatric symptomatology, weight and metabolic dysfunction in schizophrenia patients
Volume 1(1): 3-5
Clin Nutr Metab, 2018 doi: 10.15761/CNM.1000105
due to the onset of severe, high sugar food cravings for fruit, sweets and
rice. e patients showed no gastrointestinal reactions, e.g. diarrhea,
vomiting and constipation in reaction to the ketogenic diet.
Female patient
At baseline, the female patient had a calculated BMI of 21.3 kg/m2
and a measured body composition of 12.5 kg of body fat (24.5 %), 37.2
kg of muscle mass and 2 kg of bone mass. Electrocardiogram (ECG),
blood and urine analyses were clinically unremarkable and the PANSS
total score at baseline was 101 (positive = 28, negative = 16, and general
psychopathology = 57). Although, the patient failed to reach ketosis by
day 15 of the study, her calculated BMI slightly decreased to 20.7 kg/ m2
and total PANSS score reduced to 97 (positive = 28, negative = 16, and
general psychopathology = 53). Body composition remained unaltered
and outcome panels of blood and urine analyses remained clinically
unremarkable.
At conclusion of the study, the female patient had remained in
moderate/high ketosis for ~15 consecutive days and presented with a
markedly decreased calculated BMI of 19.8 kg/m2, 9.5 kg of body fat
(19.8%) and 36.6 kg of muscle mass. ere were no changes in bone
mass over the course of the study. e total PANSS score decreased to
91 (positive = 26, negative = 15, and general psychopathology = 50).
ECG results showed no alteration during this time.
Lastly, 15 days aer stopping the ketogenic diet, urine ketones were
no longer detectable. Positive and negative PANSS scores increased
to baseline initial scores, while her general psychopathology score
decreased further to 48. e calculated BMI (19.9 kg/m2) and body
composition measurements of body fat (9.9 kg) and muscle mass
(36.6 kg) slightly increased. Overall, blood clinical analyses remained
clinically unremarkable.
Male patient
At baseline, the male patient had a calculated BMI of 25.1 kg/m2 and
a measured body composition of 14.4 kg of body fat (21.7 %), 49.5 kg of
muscle mass and 2.6 kg of bone mass. Blood analysis revealed elevated
liver enzymes (AST=46 U/L & ALT=63 U/L). However, all other blood,
urine and ECG examinations remained clinically unremarkable. e
patient’s PANSS total score at baseline was 82 (positive = 19, negative
= 18, and general psychopathology = 45). At ~18 consecutive days of
moderate/high ketosis, his PANSS total score decreased to 75 (positive
= 16, negative = 17, and general psychopathology = 42) and liver
enzymes downregulated (AST=29 U/L & ALT=45 U/L), while all other
analyses of blood and urine remained clinically unremarkable.
Despite episodes of diet non-compliance at three dierent
occasions, i.e. the patient consumed rened sugars, his BMI markedly
decreased to 22.9 kg/m2, and his body fat (10.2 kg, (16.8%)) and muscle
mass (48 kg) also reduced. Although his PANSS total score increased
to 78 (positive = 17, negative = 17, and general psychopathology = 44),
it still remained below baseline testing. Interestingly, liver enzymes
normalized (AST=0-40 U/L & ALT=0-41 U/L).
Overall, there were no changes in bone mass or ECG readings over
the course of the study. 15 days aer stopping the ketogenic diet, urine
ketones were no longer detectable. Positive and negative PANSS scores
increased to baseline initial scores, while his general psychopathology
decreased further to a score of 40. e patient’s calculated BMI (23.4
kg/m2) and body composition measurements of body fat (10.9 kg)
and muscle mass (51.4 kg) slightly increased, but still remained below
baseline measurements. Overall, liver enzymes were detected at normal
levels (AST=0-40 U/L & ALT=0-41 U/L, while all other blood analyses
remained clinically unremarkable.
Conclusion
A high-fat low-carbohydrate therapeutic 3:1 ratio KD diet is a
dramatic variation from the typical Ecuadorian diet. However, its
therapeutic potential for psychiatric disorders, such as schizophrenia
merits the conduct of controlled pilot studies and clinical trials [14,15].
e underlying study evaluated the 3:1 ratio KD in a six-week pilot
study in two schizophrenia patients.
Overall, both patients showed compliance diculties with the KD
protocol and did not achieve moderate/high ketosis for prolonged times
during the study. ese warrant monitoring measures to be established
for any follow-up studies. Anticipatory planning of celebratory foods as
part of the diet may avoid a break in a future patient cohort. Similarly,
sweet cravings may be counteracted in future patient cohorts by
integrating low-sugar sweet treats, made with zero-caloric sweeteners,
e.g. stevia or erythritol, into the KD protocol.
Clinical outcomes
e gradual decrease in blood urea levels in both patients, which
was independent of a ketotic state, is inconsistent with previous
studies [29], and can potentially be explained by the patients’ reduced
dietary protein intake [30]. Blood creatinine are in line with previous
observations [31] and remained unaected throughout the study. As
liver enzymes of the female patients showed no indication of liver
abnormalities, and blood urea levels of both patients increased to
baseline measurements 15 days post-KD, the KD is not suspected to
impair liver function. Interestingly, the male patient showed elevated
liver enzymes at baseline, which could be indicative of non-alcoholic
fatty liver disease (NAFLD), or toxicity from valproic acid therapy [32-
34]. While on the KD, his liver enzymes normalized, and remained
within normal limits 15 days post-diet. Recent research suggests a KD
may improve liver enzymes and reduce triglyceride levels in hepatic
tissue [35], but other research is more ambiguous [36].
Moreover, the unaltered LDL cholesterol levels are in keeping
with results by Sharman et al. (31) of a six weeks KD in healthy, albeit
normal weight subjects. However, the underlying study failed to detect
the increased HDL cholesterol in schizophrenia patients. Despite
reports that minerals (sodium, potassium, magnesium and calcium)
can decrease with a KD in epileptic patients [10,37,38]; no alteration of
minerals in blood or bone density was exhibited during the six weeks.
However, this might be attributable to the short duration of the trial,
diet breaks and/or patient age. Lastly, the invariable ECG results across
the study are in accordance with ndings obtained by Sharma and
Gulati [39].
Body composition
Body weight, and primarily body fat, decreased over the duration
of the study, which is consistent with ndings in the eld of KD on
the whole [40-42]. Bio-impedance measurements showed a decrease in
the total muscle mass; however, this was partially reconstituted aer
two weeks of stopping the KD. e decrease in the total muscle mass
could be accounted to a depletion of the skeletal muscle glycogen
store to produce lactate [42-45], in the rst stage of the ketogenic
diet. ereaer, the citric acid function can be maintained via the
deamination of aspartate and asparagine (46), whereby preventing
skeletal tissue breakdown and ensuring the maintenance of muscle
mass. us, it appears that a KD regimen resulted in no loss of lean
muscle mass [47,48].
Gilbert-Jaramillo J (2018) e eects of the ketogenic diet on psychiatric symptomatology, weight and metabolic dysfunction in schizophrenia patients
Volume 1(1): 4-5
Clin Nutr Metab, 2018 doi: 10.15761/CNM.1000105
Psychiatric symptomatology
Our ndings in the psychiatric symptomatology in both patients
emulate the results in animal models of Krauter et al. (19), showing
improved symptomatology aer ~14 consecutive days of moderate/
high ketosis. is indicates that a short-term 3:1 ratio KD is sucient
to exert an, at least short-term, psychiatric amelioration in patients
with schizophrenia. Similarly, Ari et al. [49] suggested that a ketotic
state might be the cause of the symptom amelioration by potentially
providing a non-glucose source of energy to a potential pathological
mechanism characterized by glucose impairment and altered glucose
metabolism [50-52].
ere is an overall trend of decreasing PANSS scores during
ketosis, which is consistent with two case studies previously reported
[25]. However, by comparison the PANSS scores did not decrease to
the same extent, which could potentially be accounted to the shorter
overall duration that patients remained in ketosis. Moreover, the
general symptomatology did not return to baseline scores aer the
intervention but did showed an increased trend aer breaking the diet.
Although the PANSS interviewer remained blinded throughput the
study, post-study blinding of the examiner was compromised and could
have induced potential bias into the interpretation of the nal PANSS
interview results.
Limitations of the study
More specialized analyses e.g. change in the brain glucose uptake,
to reveal the mechanism of action of the KB in the patients, was
not available. Blood ketone beta-hydroxybutyric acid could not be
measured due to inaccessibility of laboratory protocols and devices.
is was a small pilot study intended to show a proof-of-concept,
safety, effects and overall practicability of the KD in schizophrenia
patients. Given the small sample size, statistical significance could
not be determined.
Future research
A larger sample, longer study duration is needed to determine
if these results can be extrapolated to the schizophrenia patient
population and to allow for statistical analyses to be conducted. Future
blood clinical follow-up can be done at more prolonged intervals during
a 3:1 ratio KD trial in schizophrenia patients, as no significant
changes were observed during the trial. The present protocol and
the standard laboratory assessment recommendations from ‘The
Charlie Foundation for Ketogenic Therapies’ can be used as a
template [23].
Patient perspectives
Both patients suggested that compliance with the diet daily was
dicult due to the amount of olive oil that was included in the salads,
yet, when olive oil was used for making mayonnaise, due to their culture,
made the diet easy and more exciting. Also, stop eating rice, potato,
plantains and specially sweets were accused to be the most dicult part.
Both patient reported satiety aer few days of compliance with the diet
reason why they stop having dinner 1 or 2 days. With regards to the
behavioral condition, female patient showed happiness accusing that
the two girls that regularly told to hurt herself were gone. e male
patient was feeling good when he started losing fat, recovering desire to
play football at the park with his friends. Overall, patients reported an
improved emotional condition and diet acceptance, despite most of the
time the food traditions were dicult to overcome.
References
1. Tandon R., Keshavan MS, Nasrallah HA (2008) Schizophrenia,“just the facts” what
we know in 2008. 2. Epidemiology and etiology. Schizophr res 102: 1-18. [Crossref]
2. van Os J, Kapur S (2009) Schizophrenia. Lancet 374: 635–645. [Crossref]
3. Insel TR (2010) Rethinking schizophrenia. Nature 468: 187-193. [Crossref]
4. Clay HB, Sillivan S, Konradi C (2011) Mitochondrial dysfunction and pathology in
bipolar disorder and schizophrenia. Int J Dev Neurosci 29: 311-324. [Crossref]
5. Rajasekaran A, Venkatasubramanian G, Berk M, Debnath M (2015). Mitochondrial
dysfunction in schizophrenia: pathways, mechanisms and implications. Neurosci
Biobehav Rev 48: 10-21. [Crossref]
6. Kapur S, Remington G (2001) Dopamine D 2 receptors and their role in atypical
antipsychotic action: still necessary and may even be sucient. Biol Psychiatry 50:
873-883. [Crossref]
7. Elman I, Borsook D, Lukas SE (2006) Food intake and reward mechanisms in patients
with schizophrenia: implications for metabolic disturbances and treatment with second-
generation antipsychotic agents. Neuropsychopharmacology, 31: 2091-2120. [Crossref]
8. Kang SH, Lee JI (2015) Metabolic disturbances independent of body mass in patients
with schizophrenia taking atypical antipsychotics. Psychiatry Investig 12: 242-248.
[Crossref]
9. Rado J (2017) The Complex Inter-relationship between Diabetes and Schizophrenia.
Curr Diabetes Rev. 13: 528-532. [Crossref]
10. Sampaio LPDB (2016). Ketogenic diet for epilepsy treatment. Arq Neuro-Psiquiat 74:
842-848.
11. van der Louw EJ, Williams TJ, Henry-Barron BJ, Olieman JF, Duvekot JJ, et al. (2017)
Ketogenic diet therapy for epilepsy during pregnancy: A case series. Seizure 45: 198-
201. [Crossref]
12. Stenger E, Schaeer M, Cances C, Motte J, Auvin S, et al. (2017) Ecacy of a ketogenic
diet in resistant myoclono-astatic epilepsy: A French multicenter retrospective study.
Epilepsy Res 131: 64-69. [Crossref]
13. Masino SA, Rho JM (2010) Mechanisms of ketogenic diet action. Epilepsia 51: 85-85.
[Crossref]
14. Bostock EC, Kirkby KC, Taylor BV (2017) The Current Status of the Ketogenic Diet in
Psychiatry. Front Psychiatry 8: 43 [Crossref]
15. Rho JM, Stafstrom CE (2012) The ketogenic diet as a treatment paradigm for diverse
neurological disorders. Front Pharmacol 3: 59. [Crossref]
16. Bueno NB, de Melo IS, de Oliveira SL, da Rocha AT (2013) Very-low-carbohydrate
ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised
controlled trials. Br J Nutr 110: 1178-1187. [Crossref]
17. Paoli A, Rubini A, Volek JS, Grimaldi KA (2013) Beyond weight loss: a review of
the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur J Clin Nutr 67:
789-796. [Crossref]
18. Morris AA (2005) Cerebral ketone body metabolism. J Inherit Metab Dis 28: 109-121.
[Crossref]
19. Kraeuter AK, Loxton H, Lima BC, Rudd D, Sarnyai Z (2015) Ketogenic diet
reverses behavioral abnormalities in an acute NMDA receptor hypofunction model of
schizophrenia. Schizophr Res 169: 491-493. [Crossref]
20. Palmer CM (2017) Ketogenic diet in the treatment of schizoaective disorder: Two
case studies. Schizophr Res 189: 208-209. [Crossref]
21. Pacheco A, Easterling WS, Pryer MW (1965) A pilot study of the ketogenic diet in
schizophrenia. Am J Psychiatry 121: 1110-1111. [Crossref]
22. Sánchez-Llaguno SN, Neira-Mosquera JA, Pérez-Rodríguez F, Moreno Rojas R (2013)
Preliminary nutritional assessment of the Ecuadorian diet based on a 24-h food recall
survey in Ecuador. Nutr Hosp 28: 1646-1656 [Crossref]
23. Roehl K, Sewak SL (2017) Practice Paper of the Academy of Nutrition and Dietetics:
Classic and Modied Ketogenic Diets for Treatment of Epilepsy. J Acad Nutr Diet 117:
1279-1292. [Crossref]
24. Phinney SD (2004) Ketogenic diets and physical performance. Nutr Metab (Lond) 17:
2 [Crossref]
25. Dashti HM, Mathew TC, Hussein T, Asfar SK, Behbahani A, et al. (2004) Long-term
eects of a ketogenic diet in obese patients. Exp Clin Cardiol 9: 200-205. [Crossref]
Gilbert-Jaramillo J (2018) e eects of the ketogenic diet on psychiatric symptomatology, weight and metabolic dysfunction in schizophrenia patients
Volume 1(1): 5-5
Clin Nutr Metab, 2018 doi: 10.15761/CNM.1000105
Copyright: ©2018 Gilbert-Jaramillo J. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
26. Brands MW, Manhiani MM (2012) Sodium-retaining eect of insulin in diabetes. Am J
Physiol Regul Integr Comp Physiol 303: R1101-R1109. [Crossref]
27. Urbain P, Bertz H (2016) Monitoring for compliance with a ketogenic diet: what is
the best time of day to test for urinary ketosis?. Nutr Metab (Lond) 13: 77. [Crossref]
28. Ye F, Li XJ, Jiang WL, Sun HB, Liu J (2015) Ecacy of and patient compliance with
a ketogenic diet in adults with intractable epilepsy: a meta-analysis. J Clin Neurol 11:
26-31. [Crossref]
29. Mettler S, Mitchell N, Tipton KD (2010) Increased protein intake reduces lean body
mass loss during weight loss in athletes. Med Sci Sports Exerc 42: 326-337. [Crossref]
30. Di Iorio BR, Di Micco, L, Marzocco S, De Simone E, De Blasio A, et al. (2017) Very
low-protein diet (VLPD) reduces metabolic acidosis in subjects with chronic kidney
disease: The “nutritional light signal” of the renal acid load. Nutrients 9: 69. [Crossref]
31. Sharman MJ, Kraemer WJ, Love DM, Avery NG, Gómez AL, et al. (2002) A ketogenic
diet favorably aects serum biomarkers for cardiovascular disease in normal-weight
men. J Nutr 132: 1879-1885. [Crossref]
32. Farinelli E, Giampaoli D, Cenciarini A, Cercado E, Verrotti A (2015) Valproic acid
and nonalcoholic fatty liver disease: A possible association?. World J Hepatol 7: 1251-
1257. [Crossref]
33. Verrotti A, Agostinelli S, Parisi P, Chiarelli F, Coppola G (2011) Nonalcoholic fatty
liver disease in adolescents receiving valproic acid. Epilepsy Behav 20: 382-385.
[Crossref]
34. Mnif L, Sellami R, & Masmoudi J (2016) Valproic Acid and Hepatic Steatosis: A
Possible Link? About a Case Report. Psychopharmacol bull 46: 59. [Crossref]
35. Mardinoglu A, Wu H, Bjornson E, Zhang C, Hakkarainen A, et al. (2018) An Integrated
Understanding of the Rapid Metabolic Benets of a Carbohydrate-Restricted Diet on
Hepatic Steatosis in Humans. Cell Metab 27: 559-571. [Crossref]
36. Haghighatdoost F, Salehi-Abargouei A, Surkan PJ, Azadbakht L (2016) The eects of
low carbohydrate diets on liver function tests in nonalcoholic fatty liver disease: A
systematic review and meta-analysis of clinical trials. J Res Med Sci 21: 53 [Crossref]
37. Bergqvist AC, Schall JI, Stallings VA, Zemel BS (2008) Progressive bone mineral
content loss in children with intractable epilepsy treated with the ketogenic diet. Am J
Clin Nutr 88: 1678-1684. [Crossref]
38. McDonald TJ, Cervenka MC (2017) Ketogenic Diets for Adults with Highly Refractory
Epilepsy. Epilepsy Curr 17: 346-350. [Crossref]
39. Sharma S, Gulati S (2012) The ketogenic diet and the QT interval. J Clin Neurosci 19:
181-182. [Crossref]
40. Taus M, Fumelli D, Busni D, Borroni F, Sebastianelli S, et al. (2017) A very low calorie
ketogenic diet improves weight loss and quality of life in patients with adjustable
gastric banding. Ann Ital Chir 88. [Crossref]
41. Abbasi J (2018). Interest in the Ketogenic Diet Grows for Weight Loss and Type 2
Diabetes. Jama, 319: 215-217. [Crossref]
42. Nieman DC, Carlson KA, Brandstater ME, Naegele RT, Blankenship JW (1987)
Running endurance in 27-h-fasted humans. J Appl Physiol 63: 2502-2509. [Crossref]
43. Vendelbo MH, Clasen BF, Treebak JT, Møller L, Krusenstjerna-Hafstrøm T, et
al. (2012) Insulin resistance after a 72-h fast is associated with impaired AS160
phosphorylation and accumulation of lipid and glycogen in human skeletal muscle. Am
J Physiol Endocrinol Metab 302: E190-E200. [Crossref]
44. Volek JS, Freidenreich DJ, Saenz C, Kunces LJ, Creighton BC, et al. (2016) Metabolic
characteristics of keto-adapted ultra-endurance runners. Metabolism 65: 100-110.
45. Chang CK, Borer K, Lin PJ (2017) Low-Carbohydrate-High-Fat Diet: Can it Help
Exercise Performance?. J Hum Kinet 56: 81-92. [Crossref]
46. Merra G, Miranda R, Barrucco S, Gualtieri P, Mazza M, (2016) Very-low-calorie
ketogenic diet with aminoacid supplement versus very low restricted-calorie diet for
preserving muscle mass during weight loss: a pilot double-blind study. Eur Rev Med
Pharmacol Sci 20: 2613-2621. [Crossref]
47. Gomez-Arbelaez D, Bellido D, Castro AI, Ordoñez-Mayan L, Carreira J, et al. (2016)
Body composition changes after very-low-calorie ketogenic diet in obesity evaluated
by 3 standardized methods. J Clin Endocrinol Metab, 102: 488-498. [Crossref]
48. Bertoli S, Trentani C, Ferraris C, De Giorgis V, Veggiotti P, et al. (2014) Long-term
eects of a ketogenic diet on body composition and bone mineralization in GLUT-1
deciency syndrome: a case series. Nutrition 30: 726-728. [Crossref]
49. Ari C, Kovács Z, Juhasz G, Murdun C, Goldhagen CR, et al. (2017) Exogenous ketone
supplements reduce anxiety-related behavior in Sprague-Dawley and Wistar Albino
Glaxo/Rijswijk rats. Front Mol Neurosci 9: 137. [Crossref]
50. McDermott E, de Silva P (2005) Impaired neuronal glucose uptake in pathogenesis of
schizophrenia–Can GLUT 1 and GLUT 3 decits explain imaging, post-mortem and
pharmacological ndings?. Med Hypotheses 65: 1076-1081. [Crossref]
51. Dwyer DS, Bradley RJ, Kablinger AS, Freeman AM (2001) Glucose metabolism in
relation to schizophrenia and antipsychotic drug treatment. Ann Clin Psychiatry 13:
103-113. [Crossref]
52. Pillinger T, Beck K, Gobjila C, Donocik JG, Jauhar S, et al. (2017) Impaired glucose
homeostasis in rst-episode schizophrenia: a systematic review and meta-analysis.
JAMA psychiatry 74: 261-269. [Crossref]
... Several neurological diseases involve cerebral glucose hypometabolism, which is also observed in schizophrenia; thus, it is hypothesized the ketogenic diet may be metabolically favorable for brain function in schizophrenia. There is also evidence that it can reduce oxidative stress and inflammation Ketogenic diet • Reduce oxidative stress and inflammation [28] • Compensate for impaired cerebral glucose metabolism [28] • Reduction of positive and negative symptoms [29][30][31][32][33] • Possible benefits to metabolic or gastrointestinal health [29,30] Gluten-free diet • Avoid inflammatory reactions to gluten-containing foods in patients with non-Celiac gluten sensitivity and elevated antigliadin IgG antibodies [11][12][13][14][15] • Reduction of positive and negative symptoms [34][35][36] • Improvements in cognition [35] • Possible reduction of other gluten-related symptoms, such as gastrointestinal problems Probiotics and prebiotics • Improve microbiome composition and diversity [16][17][18] • Reduction of positive and negative symptoms [38][39][40] • Improvements in cognition [42,43] • Reduction of gastrointestinal symptoms [41] Omega-3 fatty acids • Support proper neurodevelopment and neural functioning, particularly cell membranes [23] • Reduce inflammation [23] • Reduction of positive and negative symptoms [45,46] • Reduced depression and anxiety [48,60] • Improvements in cognition [58,59] Vitamin D • Modulate GABA/glutamate balance [22] • Maintain proper calcium homeostasis [22] • Reduction of positive and negative symptoms [40,63 B vitamins • Support proper neurodevelopment [19] • Support neurotransmitter formation [19] • Prevention of cognitive decline [64] • Improvements in cognition [64] and animal studies suggest it may address GABA/glutamate imbalances seen in schizophrenia [28]. While evidence on the use of the ketogenic diet in schizophrenia from human studies is limited, there are some compelling results. ...
... Several neurological diseases involve cerebral glucose hypometabolism, which is also observed in schizophrenia; thus, it is hypothesized the ketogenic diet may be metabolically favorable for brain function in schizophrenia. There is also evidence that it can reduce oxidative stress and inflammation Ketogenic diet • Reduce oxidative stress and inflammation [28] • Compensate for impaired cerebral glucose metabolism [28] • Reduction of positive and negative symptoms [29][30][31][32][33] • Possible benefits to metabolic or gastrointestinal health [29,30] Gluten-free diet • Avoid inflammatory reactions to gluten-containing foods in patients with non-Celiac gluten sensitivity and elevated antigliadin IgG antibodies [11][12][13][14][15] • Reduction of positive and negative symptoms [34][35][36] • Improvements in cognition [35] • Possible reduction of other gluten-related symptoms, such as gastrointestinal problems Probiotics and prebiotics • Improve microbiome composition and diversity [16][17][18] • Reduction of positive and negative symptoms [38][39][40] • Improvements in cognition [42,43] • Reduction of gastrointestinal symptoms [41] Omega-3 fatty acids • Support proper neurodevelopment and neural functioning, particularly cell membranes [23] • Reduce inflammation [23] • Reduction of positive and negative symptoms [45,46] • Reduced depression and anxiety [48,60] • Improvements in cognition [58,59] Vitamin D • Modulate GABA/glutamate balance [22] • Maintain proper calcium homeostasis [22] • Reduction of positive and negative symptoms [40,63 B vitamins • Support proper neurodevelopment [19] • Support neurotransmitter formation [19] • Prevention of cognitive decline [64] • Improvements in cognition [64] and animal studies suggest it may address GABA/glutamate imbalances seen in schizophrenia [28]. While evidence on the use of the ketogenic diet in schizophrenia from human studies is limited, there are some compelling results. ...
... Although these are modest changes in symptom scores, both patients still saw reductions in general psychopathology despite less-than-perfect adherence to the diet. Their scores returned to baseline 2 weeks after discontinuing the diet, but both maintained reduced body mass indices as compared to baseline [29]. In a case series describing two patients with schizophrenia diagnoses and chronic, treatment-refractory psychotic symptoms, both started a ketogenic diet for nonpsychiatric reasons (weight loss and gastrointestinal problems, respectively), but experienced complete remission of their psychiatric symptoms on the diet and were able to discontinue antipsychotics [30]. ...
Article
Full-text available
Purpose of Review This review aims to provide an overview of the pathophysiological basis for the use of nutritional strategies in the treatment of schizophrenia, outline the evidence for dietary intervention strategies, and discuss clinical considerations around their implementation. Recent Findings Inflammatory and metabolic mechanisms underlying the pathophysiology of schizophrenia are well-characterized and may provide promising treatment targets. Existing literature on dietary intervention strategies for schizophrenia provides evidence supporting the use of antiinflammatory diets, select vitamins and supplements, and more targeted approaches such as gluten-free or ketogenic diets in specific subsets of patients. Implementation of these strategies is limited by physician education on nutrition, inherent difficulties in researching nutrition, patient factors, and structural factors. Summary Nutritional approaches represent an important and potentially underutilized treatment strategy to reduce symptoms and improve quality of life for patients with schizophrenia.
... Case reports of KD abruptly resolving instances of longstanding schizophrenic symptoms provide significant hope for treatmentresistant patients (214)(215)(216)(217). Kraft and Westman (215) detailed the case of a 70-year-old woman who experienced remission of audiovisual hallucinations that had previously caused at least five episodes of hospitalization due to suicidality over the preceding 6 years despite trying lithium, olanzapine, ziprasidone, aripiprazole, quetiapine, haloperidol, lamotrigine, perphenazine, and risperidone prior to 2008. ...
Article
Full-text available
In recent times, advances in the field of metabolomics have shed greater light on the role of metabolic disturbances in neuropsychiatric conditions. The following review explores the role of ketone bodies and ketosis in both the diagnosis and treatment of three major psychiatric disorders: major depressive disorder, anxiety disorders, and schizophrenia. Distinction is made between the potential therapeutic effects of the ketogenic diet and exogenous ketone preparations, as exogenous ketones in particular offer a standardized, reproducible manner for inducing ketosis. Compelling associations between symptoms of mental distress and dysregulation in central nervous system ketone metabolism have been demonstrated in preclinical studies with putative neuroprotective effects of ketone bodies being elucidated, including effects on inflammasomes and the promotion of neurogenesis in the central nervous system. Despite emerging pre-clinical data, clinical research on ketone body effectiveness as a treatment option for psychiatric disorders remains lacking. This gap in understanding warrants further investigating, especially considering that safe and acceptable ways of inducing ketosis are readily available.
... When individuals ingest a high-fat, low-carbohydrate, and adequate protein diet, the brain defaults to utilizing ketones for energy maintenance. There is a long history of using the ketogenic diet for refractory and pediatric epilepsy (Martin et al., 2016;D'Andrea Meira et al., 2019) and more recently, this diet has been studied in serious mental illness (Kraft and Westman, 2009;Bostock et al., 2017;Gilbert-Jaramillo et al., 2018). Neuroimaging studies have found that the increased neurometabolic efficiency of ketosis translates to greater network stability, as measured by sustained functional communication between regions from BOLD fMRI (Mujica-Parodi et al., 2020). ...
Article
Full-text available
The brain is a living organ with distinct metabolic constraints. However, these constraints are typically considered as secondary or supportive of information processing which is primarily performed by neurons. The default operational definition of neural information processing is that (1) it is ultimately encoded as a change in individual neuronal firing rate as this correlates with the presentation of a peripheral stimulus, motor action or cognitive task. Two additional assumptions are associated with this default interpretation: (2) that the incessant background firing activity against which changes in activity are measured plays no role in assigning significance to the extrinsically evoked change in neural firing, and (3) that the metabolic energy that sustains this background activity and which correlates with differences in neuronal firing rate is merely a response to an evoked change in neuronal activity. These assumptions underlie the design, implementation, and interpretation of neuroimaging studies, particularly fMRI, which relies on changes in blood oxygen as an indirect measure of neural activity. In this article we reconsider all three of these assumptions in light of recent evidence. We suggest that by combining EEG with fMRI, new experimental work can reconcile emerging controversies in neurovascular coupling and the significance of ongoing, background activity during resting-state paradigms. A new conceptual framework for neuroimaging paradigms is developed to investigate how ongoing neural activity is “entangled” with metabolism. That is, in addition to being recruited to support locally evoked neuronal activity (the traditional hemodynamic response), changes in metabolic support may be independently “invoked” by non-local brain regions, yielding flexible neurovascular coupling dynamics that inform the cognitive context. This framework demonstrates how multimodal neuroimaging is necessary to probe the neurometabolic foundations of cognition, with implications for the study of neuropsychiatric disorders.
... [121] Clinical studies have suggested that KDs are effective in treating schizophrenia. [122,123] In MK801-treated mice, acute 3HB attenuated spontaneous motor activity, MK801-induced motor hyperactivity, and MK801-induced prepulse inhibition. [124] 3HB offered a new treatment option for individuals with schizophrenia by 6 of 10 -WANG ET AL. ...
Article
Full-text available
3‐hydroxybutyrate (3HB), or BHB, is an anionic small molecule acid metabolite with a hydroxyl group. 3HB is the major ketone body that is distributed in the human brain and its primary energy source when glucose is absent. A better understanding of 3HB and how to adapt neuronal response mechanisms is expected to facilitate the development of new interventions to promote cognitive brain function and prevent neurodegenerative diseases. It provides important concepts for the clinical application of 3HB therapy. This review summarizes the distribution of 3HB in the brain, its properties, and its mechanism in brain and nerve regulation. We focus on 3HB biosynthesis in natural human cells and engineered bacteria via synthetic biology platforms and 3HB treatment in various brain and nerve diseases, including epilepsy, multiple sclerosis, stroke, Parkinson's disease, Alzheimer's disease, Huntington's disease, depressive disorder, and schizophrenia. Ultimately, this review explores the future development trend of 3HB as a potential small‐molecule drug for brain and nerve diseases.
Article
Coordinated regulation of energy conversion processes in the brain maintains its highly productive work and efficient mental activity. Impairments of the brain energy metabolism are considered among pathogenetic factors in the schizophrenia origin, but presently it is difficult to say whether these impairments are primary and causative the development of the disease or represent consequences of certain changes in the functioning of neurotransmitter and other neurochemical systems. This review discusses the main results of the energy metabolism research in schizophrenia – at various levels and using different approaches, as well as regards some attempts of influencing the energy processes in the brain as an adjunctive therapy in schizophrenia. To date, the efficacy of these therapeutic approaches has not been proven, this may be due to the paucity of studies and the lack of preliminary identification/stratification of patient subgroups to whom the energy metabolism-targeted therapy would be the most useful. Based on the data presented, one can conclude that an analysis is necessary of relationships between the psychopathological manifestations of schizophrenia and energy metabolism deviations for further identification of those patients to whom the use of mitochondrial modulators, mitoprotection, and other approaches may represent a promising method of adjunctive therapy.
Article
Full-text available
Ketogenic diet reduces pathological stress and improves mood in neurodegenerative and neurodevelopmental disorders. However, the effects of ketogenic diet for people from the general population have largely been unexplored. Ketogenic diet is increasingly used for weight loss. Research in healthy individuals primarily focuses on the physical implications of ketogenic diet. It is important to understand the holistic effects of ketogenic diet not only the physiological but also the psychological impacts in non-clinical samples. The aim of this cross-sectional study with multiple cohorts was to investigate the association of ketogenic diet with different aspects of mental health including calmness, contentedness, alertness, cognitive and emotional stress, depression, anxiety and loneliness in a general healthy population. Two online surveys were distributed: Cohort 1 using the Bond-Lader visual analogue scale (BL-VAS) and Perceived Stress Scale (PSS-10) (n=147) and Cohort 2 the Depression, Anxiety and Stress Scale (DASS-21) and UCLA-R Loneliness scale (n=276). Ketogenic diet was associated with higher self-reported mental and emotional well-being behaviours including calmness, contentedness, alertness, cognitive and emotional stress, depression, anxiety and loneliness compared to individuals on a non-specific diet in a general population. This research demonstrated that ketogenic diet has potential psychological benefits within the general population.
Article
Schizophrenia is a severe illness that affects 24 million individuals around the globe. Individuals with schizophrenia have been known to make poor nutritional choices. In fact, they are known to consume a lot more sugar and fat, and less fruits and vegetables. In this paper, I show how diet can reduce the symptoms of schizophrenia; that is, that consumption of certain foods can lessen the symptoms of schizophrenia. Further, I create an optimal diet for individuals with schizophrenia.
Article
Full-text available
As perturbações mentais estão a tornar-se mais prevalentes a nível global, e existe evidência de que a nutrição é um fator crucial no que diz respeito à sua prevalência e incidência. As abordagens de tratamento mais utilizadas, a psicoterapia e a psicofarmacoterapia, nem sempre são acessíveis, toleráveis ou eficazes no alívio dos sintomas. As dietas cetogénicas, restritas em hidratos de carbono, moderadas em proteína e ricas em gordura, são há muito utilizadas no tratamento de epilepsia refratária e, mais recentemente, têm vindo a ser investigadas no âmbito de muitas outras condições metabólicas, neurodegenerativas e do neurodesenvolvimento. Este trabalho pretende rever a evidência existente relativamente à utilização das dietas cetogénicas na prevenção e tratamento de perturbações mentais. Foi realizada uma pesquisa nas bases de dados PubMed e Scopus, de fevereiro a junho de 2022. Os estudos encontrados atribuem às dietas cetogénicas um efeito ansiolítico, antidepressivo, estabilizador do humor e antipsicótico, para além de reduções significativas na sintomatologia de várias outras perturbações, como autismo, ingestão compulsiva, hiperatividade e abuso de substâncias. Contudo, os estudos em humanos são escassos e de baixa qualidade. Em conclusão, a escassez de ensaios clínicos aleatorizados e controlados não permite traçar, neste momento, conclusões firmes sobre a eficácia das dietas cetogénicas nas perturbações mentais. No entanto, o seu carácter promissor justifica a realização de mais estudos.
Article
Full-text available
Schizophrenia is a mental illness that manifests itself in the second or third decade of life with no coherent aspect. The primary pharmacological therapy is antipsychotic medication, which mostly work by suppressing the activity of dopamine. Unluckily, many of schizophrenic patients experience hardship from severe positive or negative symptoms that can not be completely treated with available treatment. Last theory of schizophrenia mechanism contain systemic and central proinflammatory process, in brain glutamatergic neurotransmission and impaired systemic and cerebral energy metabolism. Ketogenic diet is firm controlled high-fat, low protein, low-carbohydrate diet, with lipid to non lipid ratio 4:1. Main mechanism is to cause ketosis. The situation in which ketones bodies are used as a substrate to generate energy in the organism. It could be that a ketogenic diet could help to compensate for the imbalance of GABA levels in schizophrenic brain. The ketogenic diet as an addition to drug treatment, hopefully may offer therapeutic opportunities to better result of the disease concerning symptomatology and preventing the gain of weight, due to some medication side effects or comorbidities.
Article
Full-text available
Background: Valproic acid is a mood-stabilizing anticonvulsant. Hepatic injuries are among the occasionally observed adverse effects of this medication. Case presentation: We present the case of a 47-year-old man who had bipolar disorder for ten years and treated with valproic acid. He demonstrated elevated serum aminotransferases and ultrasonography revealed that hepatomegaly was suggestive of hepatic steatosis. Conclusion: This case report stresses the importance of a complete drug history and the need for clinicians to be aware of the delayed onset of hepatic injuries.
Article
Full-text available
Low-carbohydrate-high-fat (LCHF) diets have been used as a means of weight loss and control of symptoms in several clinical conditions. There is emerging evidence that the metabolic changes induced by LCHF diets enhance endurance performance. The aims of this review are to examine the evidence of LCHF diets in improving various aspects of athletic performance. Long-term LCHF dietary intake may help control body weight and fat mass while maintaining lean body mass in athletes in weight-sensitive sports. LCHF-adapted endurance athletes can reach the maximal fat oxidation rate of approximately 1.5 g/min, with a lower carbohydrate oxidation rate and similar muscle glycogen content and a resynthesis rate compared to their counterparts consuming high-carbohydrate-low-fat (HCLF) diets. The elevated fat oxidation rate and glycogen sparing effect may improve performance in ultra-endurance events. These metabolic changes may also prevent the decline in performance in later stages of repeated high-intensity movements, in which the aerobic metabolism becomes more important. However, elevated blood concentrations of non-esterified fatty acids and ammonia during exercise after LCHF diets may lead to early development of central fatigue. It appears that at least several months of adaptation to a LCHF diet are required for the metabolic changes and restoration of muscle glycogen to occur. Further investigations on LCHF diets are needed regarding (1) performance after weight loss in weight-categorized sports; (2) repeated high-intensity exercise performance; (3) development of central fatigue during endurance events; (4) perceptual-motor performance during prolonged intermittent sports; and (5) ideal dietary fatty acid compositions.
Article
Full-text available
Background The ketogenic diet (KD) has been used in treatment-resistant epilepsy since the 1920s. It has been researched in a variety of neurological conditions in both animal models and human trials. The aim of this review is to clarify the potential role of KD in psychiatry. Methods Narrative review of electronic databases PubMED, PsychINFO, and Scopus. Results The search yielded 15 studies that related the use of KD in mental disorders including anxiety, depression, bipolar disorder, schizophrenia, autism spectrum disorder (ASD), and attention deficit hyperactivity disorder (ADHD). These studies comprised nine animal models, four case studies, and two open-label studies in humans. In anxiety, exogenous ketone supplementation reduced anxiety-related behaviors in a rat model. In depression, KD significantly reduced depression-like behaviors in rat and mice models in two controlled studies. In bipolar disorder, one case study reported a reduction in symptomatology, while a second case study reported no improvement. In schizophrenia, an open-label study in female patients (n = 10) reported reduced symptoms after 2 weeks of KD, a single case study reported no improvement. In a brief report, 3 weeks of KD in a mouse model normalized pathological behaviors. In ASD, an open-label study in children (n = 30) reported no significant improvement; one case study reported a pronounced and sustained response to KD. In ASD, in four controlled animal studies, KD significantly reduced ASD-related behaviors in mice and rats. In ADHD, in one controlled trial of KD in dogs with comorbid epilepsy, both conditions significantly improved. Conclusion Despite its long history in neurology, the role of KD in mental disorders is unclear. Half of the published studies are based on animal models of mental disorders with limited generalizability to the analog conditions in humans. The review lists some major limitations including the lack of measuring ketone levels in four studies and the issue of compliance to the rigid diet in humans. Currently, there is insufficient evidence for the use of KD in mental disorders, and it is not a recommended treatment option. Future research should include long-term, prospective, randomized, placebo-controlled crossover dietary trials to examine the effect of KD in various mental disorders.
Article
A carbohydrate-restricted diet is a widely recommended intervention for non-alcoholic fatty liver disease (NAFLD), but a systematic perspective on the multiple benefits of this diet is lacking. Here, we performed a short-term intervention with an isocaloric low-carbohydrate diet with increased protein content in obese subjects with NAFLD and characterized the resulting alterations in metabolism and the gut microbiota using a multi-omics approach. We observed rapid and dramatic reductions of liver fat and other cardiometabolic risk factors paralleled by (1) marked decreases in hepatic de novo lipogenesis; (2) large increases in serum β-hydroxybutyrate concentrations, reflecting increased mitochondrial β-oxidation; and (3) rapid increases in folate-producing Streptococcus and serum folate concentrations. Liver transcriptomic analysis on biopsy samples from a second cohort revealed downregulation of the fatty acid synthesis pathway and upregulation of folate-mediated one-carbon metabolism and fatty acid oxidation pathways. Our results highlight the potential of exploring diet-microbiota interactions for treating NAFLD.
Article
This summer, 25 overweight and obese adults participating in a tightly controlled feeding study will take up full-time residence for 3 months at a wooded lakefront center in Ashland, Massachusetts. However, before checking in at Framingham State University’s Warren Conference Center and Inn, they will have to lose 15% of their body weight on a calorie-restricted diet with home-delivered meals.
Article
The current review highlights the evidence supporting the use of ketogenic diets in the management of drug-resistant epilepsy and status epilepticus in adults. Ketogenic diet variants are compared and advantages and potential side effects of diet therapy are discussed.
Article
Ketogenic diet (KD) therapy is an established form of treatment for both pediatric and adult patients with intractable epilepsy. Ketogenic diet is a term that refers to any diet therapy in which dietary composition would be expected to result in a ketogenic state of human metabolism. While historically considered a last-resort therapy, classic KDs and their modified counterparts, including the modified Atkins diet and low glycemic index treatment, are gaining ground for use across the spectrum of seizure disorders. Registered dietitian nutritionists are often the first line and the most influential team members when it comes to treating those on KD therapy. This paper offers registered dietitian nutritionists insight into the history of KD therapy, an overview of the various diets, and a brief review of the literature with regard to efficacy; provides basic guidelines for practical implementation and coordination of care across multiple health care and community settings; and describes the role of registered dietitian nutritionists in achieving successful KD therapy.
Article
Background: Often, in severe obesity, diet and physical activity are not enough to achieve a healthy BMI. Bariatric surgical approach, in particular laparoscopic adjustable gastric banding (LAGB), has encouraging results in terms of weight loss and resolution of obesity-related comorbidities. However, several months after LAGB, some patients are enable to lose weight anymore and don't tolerate a further calibration because of its collateral effects (excessive sense of fullness, heartburn, regurgitation and vomiting). Aim: The aim of this study is to identify the potential role of high protein-low carbohydrate ketogenic diet (KD) in managing weight loss in patients who underwent gastric banding and didn't lose weight anymore. Methods: 50 patients underwent LAGB between January 2010 and December 2013. In twenty patients (GROUP A) we observed a stop in weight loss so we divided this patients into two groups. One group (group A1: 10 patients) continued to follow a LCD low calorie diet and underwent a further calibration; the other group (group A2: 10 patients) started to follow a KD for the next 8 weeks. Results: Both group resumed a significant weight loss, however group A1 patients reported collateral effects due to calibration and a higher Impact of Weight on Quality of Life - Lite (IWQOL-Lite) that correlates with a lower quality of life than patients following KD. Conclusions: KD can improve the weight loss and quality of life in patients who underwent LAGB and failed at losing more weight allowing a weight loss comparable to that obtained with a further calibration and it is useful to avoid drastic calibrations and their collateral effects. Key words: Laparoscopic adjustable gastric binding, Quality of life, Very low calory ketogenic binding.