MINI REVIEW article

Front. Nutr., 10 March 2025

Sec. Nutrition, Psychology and Brain Health

Volume 12 - 2025 | https://doi.org/10.3389/fnut.2025.1492306

This article is part of the Research TopicKetogenic Metabolic Therapy as a Treatment for Mental Health DisordersView all 15 articles

Ketogenic diet, adenosine, and dopamine in addiction and psychiatry

  • Trinity College, Hartford, CT, United States

Adhering to the ketogenic diet can reduce or stop seizures, even when other treatments fail, via mechanism(s) distinct from other available therapies. These results have led to interest in the diet for treating conditions such as Alzheimer’s disease, depression and schizophrenia. Evidence points to the neuromodulator adenosine as a key mechanism underlying therapeutic benefits of a ketogenic diet. Adenosine represents a unique and direct link among cell energy, neuronal activity, and gene expression, and adenosine receptors form functional heteromers with dopamine receptors. The importance of the dopaminergic system is established in addiction, as are the challenges of modulating the dopamine system directly. A mediator that could antagonize dopamine’s effects would be useful, and adenosine is such a mediator due to its function and location. Studies report that the ketogenic diet improves cognition, sociability, and perseverative behaviors, and might improve depression. Many of the translational opportunities based on the ketogenic diet/adenosine link have come to the fore, including addiction, autism spectrum disorder, painful conditions, and a range of hyperdopaminergic disorders.

Introduction

Metabolic therapy with the ketogenic diet (KD) has been used successfully to treat epilepsy in adults and children for over 100 years (1). Adhering to this high-fat, low-carbohydrate protocol can reduce or even stop seizures – even when all other treatments fail, and some pediatric patients are able to discontinue the KD and remain seizure-free (26). This effect is also found with laboratory animals (7), indicating a disease-modifying, antiepileptogenic effect found only weakly in some but not present at all in most anticonvulsant medications (8, 9). These observations indicate that this metabolic therapy works via mechanism(s) distinct from other available therapies—and demonstrate clinically that a KD may permanently restore normal brain function.

The proven, long-term efficacy in epilepsy has led to interest in the KD’s mechanisms for preventing and treating multiple conditions (such as diabetes), but particularly in other neurological conditions such as Parkinson’s disease, Alzheimer’s disease, and multiple sclerosis (10), as well as conditions where seizures are often comorbid. Several types of behavioral evidence predict benefits of a KD in reducing common comorbidities other than seizures such as depression (11, 12) and anxiety (11, 13). Most studies report that the KD improves cognition (1319), improves sociability and repetitive behaviors (2023), and reduces nociception (2426): all behavioral endpoints with relevance to dopamine-related behaviors (see below), including perseverative behaviors and potentially chronic pain – thought to share multiple mechanisms and comorbidities with addiction (27). Importantly, KD-related behavioral improvement in children with epilepsy is not solely due to seizure reduction (13, 2832), thus indicating therapeutic benefits that are uncorrelated with the primary anticonvulsant/antiepileptogenic effects.

Diverse lines of evidence point to the neuromodulator adenosine as a key mechanism underlying short and long-term therapeutic benefits of metabolic therapy with a KD. Adenosine is present throughout the extracellular space, and its levels increase with increased neural activity (33, 34) and a variety of physiological conditions (35). We put forth this hypothesis and its translational predictions in 2008 (36). Since then, we developed metabolic models and provided in vitro and in vivo evidence that KD feeding elevates brain adenosine (7, 3639). More evidence has since accumulated (4042), and many of the translational opportunities based on the KD/adenosine hypothesis have come to the fore, including pain, autism spectrum disorder, neuroprotection, and a range of hyperdopaminergic disorders (35, 36). Adenosine represents a unique and direct link among cell energy, neuronal activity, and gene expression and a direct functional relationship with dopamine. Here, we review several molecular/physiological actions of the KD by which the KD might influence addiction and psychiatric disorders, then delve into specific disorders with respect to KD treatment.

Adenosine/dopamine interactions

The behavioral importance of the dopaminergic system is well-established – as are the challenges and limitations (side effects, limited therapeutic windows) of modulating the dopamine system directly. Dopamine release is clearly related to the reinforcing effect of drugs of abuse, such as cocaine, which blocks re-uptake of dopamine and so increases extracellular levels of this neurotransmitter. The discussion below is largely focused on cocaine. Chronic use of this drug in people leads to a number of behavioral sequelae, including highly-motivated use even in the face of adverse consequences. Laboratory rodents chronically self-administering cocaine show similar behaviors (43), including no diminution of self-administration even in the face of a signal of impending footshock (43, 44). Remarkably, cocaine cravings increase over 60 days of withdrawal in rodents (45), in accordance with reports in human addiction. PET studies in cocaine-addicted patients show reduced D2 dopamine receptor levels in the basal ganglia and reduced metabolism in the cingulate gyrus and orbitofrontal cerebral cortex (46). Brain effects of cocaine progress with extended self-administration, with extension of metabolic changes from the limbic basal ganglia to the entire basal ganglia in Rhesus monkeys (47), and progressively elevated levels of brain-derived neurotrophic factor in the limbic basal ganglia and amygdala; this protein causes long-lasting amplification of cocaine seeking (48).

A mediator that could interfere with the effects of dopamine (without blocking it completely) would be extremely useful, and adenosine is such a mediator due to its function and its location. Manipulating the adenosine system is common – caffeine, a non-selective antagonist for adenosine A1 receptors (A1R) and adenosine A2 receptors (A2R), is the most widely used psychoactive drug worldwide – and other adenosine antagonists are under consideration for neurodegenerative and psychiatric disorders (49, 50). Notably, subpopulations of richly dopamine-innervated basal ganglia neurons express either a combination of A1R and D1 dopamine receptors (D1R) or A2AR and D2 dopamine receptors (D2R) (51, 52), and these colocalized receptors form functional heteromers with antagonistic effects on 2nd messenger systems (5358).

These oppositional relationships also appear at the behavioral level in rodents, in work often involving cocaine. For example, A2aR agonists decreased, whereas A2aR antagonists increased, acute cocaine-induced locomotion, in apparent opposition to the D2R (57). Selectively knocking out A2aR expression in striatal neurons enhances the locomotor response to cocaine or phencyclidine (58). Chronic caffeine in adolescence increases the locomotion to a challenge dose of cocaine or a D2R agonist in adulthood (59). Outside the brain, A1R and D1R oppositely influence spinal motor circuit output (60). Caffeine reduced the locomotor sensitization response to cocaine in a binge protocol (61). Given during a sensitization regimen, A2aR agonists decreased, whereas A2aR antagonists increased, the sensitized response to a later cocaine challenge (57). Alternatively, A1R or A2aR agonists given during the cocaine challenge but not during sensitization reduced the expression of cocaine sensitization, in a paradigm in which the adenosinergic drugs were directly infused into the basal ganglia (62).

In the conditioned place preference test, adolescent chronic caffeine enhanced the rewarding effect of cocaine in adulthood (59). An A2aR agonist reduced the reinforcing and motivational aspects of cocaine self-administration (63). A1R agonists inhibited cocaine- or D1R agonist-induced reinstatement of extinguished cocaine self-administration (64). Caffeine potentiated the seizure-inducing properties and lethality of cocaine and D-amphetamine (65). Also relevant to drug abuse, adenosine and dopamine (mostly the A2aR and D2R) differentially control motivation (66). Overall, there is an abundance of evidence that adenosine and dopamine receptors are in opposition in their influence on several types of behavior and cognition.

Some evidence suggests the KD alters dopamine directly. The dopamine metabolite homovanillic acid was reduced during KD feeding in pediatric epileptic patients in a study that used CSF as a proxy for tissue dopaminergic activity, though this effect did not differ with presence or absence of anticonvulsant response (67). In rats, tissue homovanillic acid (combined with another dopamine metabolite, dihydroxyphenylacetic acid) was elevated by the KD in cerebral cortex but not basal ganglia or midbrain (68). These differences could be explained by a number of factors, such as species differences, differences in subject maturity, differences in KD strength/composition, or the effective whole-brain sampling of CSF collection. A KD-based mechanism to moderate adenosine and/or dopamine systems would have obvious relevance to neurological conditions, including drug abuse (69, 70).

Cerebrocortical hypometabolism versus energy replenishment

Hypotheses and clinical and basic research on the link between brain energy and mental health has been a rapidly developing field with case reports, reviews, protocols, and cutting-edge conferences helping to foster a robust and thriving community with real collaboration between patients and professionals (7176). Compensating for the energy impairment due to ongoing hypometabolism may be a useful treatment for many diverse neurological conditions (7780). Energy homeostasis – particularly changes in ATP and adenosine – is known to be relevant but poorly understood in neuroprotection, psychiatric disorders and addiction (81, 82). KDs supply a substrate (ketone bodies) for the citric acid cycle that elevates ATP and promotes mitochondrial function, including in impaired states (8397).

Brain hypometabolism has been reported with alcohol and online gaming addiction (98), with stimulant abuse (99, 100), in Alzheimer’s disease and mild cognitive impairment (101, 102), and indeed even with normal aging (103). As a dynamic and energy-demanding organ, and as a survival mechanism, it makes sense that metabolism is reflected in neurological function and behavior and that mitigating metabolic dysfunction is a potent therapeutic strategy.

Reduced hyperglycemia and/or inflammation

KDs produce a moderately low but very stable blood glucose (104107), explaining why it is an effective treatment for diabetes (106, 108, 109). This stabilization of blood glucose may blunt the impact of well-known physiological effects of stress and/or dopamine-induced hyperglycemia (110), and therefore may help stabilize a range of mental states that are influenced by metabolic variability, particularly those that are triggered by or associated with hyperglycemia. Hyperglycemia causes inflammation (111, 112) and is associated with psychiatric re-hospitalization. Inflammation is a biomarker for and perhaps a cause of depression (113), and much evidence shows that KDs reduce inflammation in patients (114117) and in pre-clinical models (118121). Importantly, some animal studies found reduced inflammatory markers specifically in brain (122125). KD feeding seems to limit neuroinflammation via several mechanisms (126).

Disorders

Addiction

Based on the relationships among adenosine, dopamine, and the KD, we recently investigated the possible moderating effects of KD treatment on the effects of repeated cocaine treatments (127). Five-week old male and female rats were placed on a KD or remained on normal rodent chow for 3 weeks. A well-established cocaine-sensitizing regimen was then applied: animals received once-daily injections of either saline or cocaine for seven consecutive days, followed by seven drug-free days, and then finally a challenge injection of cocaine. Assessments occurred in an automated system for measuring ambulatory (e.g., walking) and stereotyped (e.g., rearing) locomotor responses. KD feeding continued through the sensitization protocol. All animals receiving the daily cocaine injections showed the expected enhancement of the rearing response, but animals on the KD had a significantly mitigated enhancement. Unexpectedly, ambulatory activity did not sensitize at all in KD-fed animals. These effects of KD on locomotor activity were found in both sexes, and were only observed following injections of cocaine (not saline). A similar pattern was found with the challenge injection: KD treatment moderated the stereotypic response to the challenge. Interestingly, here sex was a factor, with this effect occurring in males only. Thus, KD feeding reduces both the responses to acute cocaine (day one of the sensitizing regimen; challenge day for saline-treated animals) and repeated cocaine. Considered together, these data were the first to show that KD treatment can modify behavioral responses to a monoaminergic stimulant, and suggest that KDs are a potential novel therapy for the treatment of addiction to these drugs. Based on prior studies, we posit that the effects of the KD in this paradigm could be mediated by an effect of adenosine on dopaminergic systems, likely in the basal ganglia.

More recently, the effects of KD treatment were tested in a conditioned place preference protocol, wherein animals learn to prefer a section of the experimental apparatus paired with, in this case, cocaine injections (128). KD feeding did not appear to modulate the acquisition of the cocaine-related place preference. However, when cocaine was withheld (i.e. extinction), mice on the KD more quickly lost the place preference. In addition, a cocaine priming injection after extinction reinstated the place preference only for the standard diet mice; mice that were on KD did not experience reinstatement. The authors hypothesized that the KD effects were via an adenosine/dopamine interaction, and suggested that KD treatment might be especially useful in preventing relapse.

Regarding the commonly abused drug ethanol, in rat models of dependence KD-fed animals made fewer lever presses to receive alcohol during acute withdrawal (129) and had reduced withdrawal symptoms (130, 131). In mice, both KD and a ketone monoester (which is metabolized to ketone bodies) reduced withdrawal symptoms even when treatment was started during withdrawal (132). Clinically, benzodiazepines are given to reduce withdrawal symptoms during detoxification: notably, patients eating a KD during treatment required significantly fewer or lower doses of benzodiazepines (129). Alcohol-related stimuli induced fewer or lower doses of “wanting” and more dorsal anterior cingulate gyrus activation in patients on a KD; neuroinflammatory markers were also reduced (129). An alcohol-dependent metabolism has switched from depending on glucose to depending on acetate; ketone bodies might normalize metabolism by replacing acetate (133). It had been hypothesized that alcohol addiction might relate to adenosine dysfunction in the basal ganglia (134) and a recent study provides direct evidence (135).

Adenosine is clearly involved in the effects of opiates. During a dependence-inducing regimen of morphine and during withdrawal, brainstem adenosine was reduced two-fold (136). During withdrawal, symptoms were reduced with an A1R agonist or an A2aR antagonist (136) or genetic inactivation of A2aR (137). Consistent with these results, KD feeding reduced symptoms of withdrawal from opiates in mice (138, 139). In addition, KD feeding reduced opiate self-administration (139) and hyperalgesia due to chronic opiate treatment (140). These results suggest a KD, through adenosine, might have some utility in opiate abuse. Conversely, a KD elevated locomotor responses and analgesia to oxycodone. This latter effect, however, could be partially explained by the antinociception due to the KD itself (25, 120).

Food cravings, binge eating being an extreme form, are often considered to be a naturalistic analog of drug abuse. Excessive glucose and insulin spikes are thought to modify the brain leading to addiction-like binge eating; KD feeding will temper such spikes (141). Two pilot studies of KD treatment to patients with food addiction/binge eating disorder underwent KD treatment, leading to significant reductions (142) or complete alleviation (143) of the disorder’s symptoms.

Psychiatric disorders

A recent study found that KD treatment in 28 patients with severe refractory mental illness significantly improved psychotic symptoms and depression; virtually every patient improved on multiple scales (144). Twelve of the patients achieved clinical remission on the Clinical Global Impressions Scale. A majority of patients reduced number or dose of psychotropic medications (in a number of cases, diabetes-related medications were reduced or discontinued) (144). After discharge, 18 patients chose to remain or partially remain on the diet to maintain the psychiatric benefits. Subsequent studies have also found broad KD effectiveness in mental illness such as bipolar disorder and schizophrenia (145151). Much evidence shows that KDs reduce inflammation in patients (114117, 152) and in pre-clinical models (118121). Reductions in inflammation might be particularly germane to depression (113).

An involvement of adenosine (specifically, an alteration in normal adenosine/dopamine antagonism) has long been postulated for schizophrenia (153155), and adenosine modulators have been tried with some success in patients (156). More recent papers have highlighted abnormalities in adenosine receptor expression specifically in frontal cerebral cortex but not other adenosine receptor expressing regions (157, 158). In parallel, hypometabolism, limited to the frontal cerebral cortex, was indicated in schizophrenia by meta-analysis (159). Therefore, the KD might have beneficial effects via multiple mechanisms. One group found positive effects in an animal model of schizophrenia-like behavior (160162). A very early attempt to use the KD in schizophrenic patients showed promise but was poorly controlled (163). More recently, beneficial results have been reported, but these are either case reports (164, 165) or have a low number of subjects (five schizophrenic or schizoaffective patients) (151). Larger studies are warranted, although in a study with a substantial sample size the KD reduced schizotypy traits in the general population (166).

Relating to hyperglycemia, diabetes is associated with a higher incidence of several mood and psychiatric disorders (167, 168). A meta-analysis found a significant association between depression and both type I and type II diabetes (169). In diabetic individuals, hyperglycemia is associated with depression (170) and feelings of anger and sadness (171), which may be worse in type I diabetes (170). Such effects are not limited to diabetic patients: hyperglycemia is related to higher readmittance to psychiatric hospitalization (172), and high insulin levels in youth raise the odds of psychosis in young adulthood (173). On more acute timescales, there is some evidence for high glycemic variability relating to low quality of life and negative mood in diabetic patients (174176), although other studies have not found support for this association (177, 178). Notably, high dietary sugar intake is associated with depression and anxiety in non-diabetic individuals (179181). These associations do not determine causation but, intriguingly, there are suggestions that depression in the elderly might predispose the development of type II diabetes (182, 183). KDs minimize dietary sugar intake, and provided a stable, mild hypoglycemia which should counteract these deleterious effects on mood. A recent review outlined the heightened risk of dementia in type II diabetes, and the use of KDs as a preventative treatment (184).

Cerebral hypometabolism/hypoperfusion is known to factor into cognitive problems in Alzheimer’s disease, dementia, and mild cognitive impairment; ketogenic strategies can overcome this problem by delivering high energy fuels (ketone bodies) directly to neural tissue (82). A recent report showed that KD or ketone body treatment restores long-term potentiation in a mouse model of Alzheimer’s disease (185). A number of clinical studies have applied the KD (or the modified Atkins diet, also very low carbohydrate) to these disorders (186). Although cognitive tests differ between study groups, the KD is generally found to benefit general cognition, learning and memory, quality of life, general functioning, and mood (187191). In one study, serum ketones were found to positively correlate with benefits in long term memory (17). Other studies have more directly induced ketosis in these patients with supplements, typically medium-chain triglycerides or ketone esters (which are easily metabolized into ketone bodies), rather than changing diet wholesale. Again, these treatments improved various aspects of cognition (77, 192). A number of studies correlated improved cognition with elevated circulating ketone bodies (193195) or elevated ketone body uptake in brain (assessed with PET) (196, 197).

There is strong evidence of a metabolic underpinning of ASD, in addition to the genetic and environmental components. For example, this disorder has been found to involve hypoperfusion of specific brain regions (198200) and to be associated with hyperglycemia, mitochondrial dysfunction, and adenosine dysfunction (201204). Thus, the KD has multiple mechanisms by which it might be beneficial. KD feeding improved sociability and repetitive behaviors in various animal models of this disorder (2628, 91, 205, 206). In addition, promising results have been found in autism spectrum disorder patients with KD therapy (207213).

Addiction and the psychiatric disorders just discussed all have significant co-morbidities; interestingly, KD treatment appears to be helpful with many of these co-morbidities. Diabetes is a co-morbidity in depression, schizophrenia, and Alzheimer’s disease; KD feeding is a greatly beneficial treatment for diabetes (214). Obesity is a co-morbidity in schizophrenia and Alzheimer’s disease; KD feeding is an effective treatment for obesity (215). Attention deficit/hyperactivity disorder is a co-morbidity in addiction and autism spectrum disorder; KD feeding improves attention during treatment of epileptic patients (31, 216218). Hyperactivity was improved during treatment in epilepsy and autism spectrum disorder (210, 219), but has not been established as a treatment in a non-epileptic clinical ADHD population. Depression is a co-morbidity in addiction and Alzheimer’s disease; KD feeding is effectively antidepressant in non-epileptic populations (11, 117, 144). For rarer co-morbidities such as personality disorder, KD effects remain unknown.

The KD can have some effects on lipids which can be seen as possible negative side effects. However, mild hyperlipidemia was associated with better anticonvulsant effects (219). Even though low-density lipoprotein-C was higher in KD-fed patients, there was no increased coronary plaque burden compared to matched controls (220), and low-density lipoprotein-C levels are generally poorly predictive of cardiovascular disease risk (221, 222).

Taken together, the relationship between metabolic health and a broad range of neurological conditions is emerging, including mental health. The relationship among adenosine, dopamine, and ketogenic metabolic therapy is primary because of the ability to link cell energy, neuronal signaling, and gene expression for both short and long-term effects in key brain areas. The opportunity for metabolic approaches to address multiple comorbidities at once is gaining acceptance. As noted herein there is a wide range of mechanisms and impacts, and reversing and preventing metabolic dysfunction has enormous potential for all ages. However, the opportunity to restore a lifetime of brain health for young people – who may be suffering from mental illness, drug addiction, or both – should be an enormous motivation for continued attention to this field.

Author contributions

DR: Conceptualization, Writing – original draft, Writing – review & editing. LM: Investigation, Writing – review & editing. SM: Writing – review & editing, Conceptualization, Supervision, Writing – original draft.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by NIH NS065597 (SAM) and AT008742 (DNR).

Conflict of interest

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

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Publisher’s note

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

References

1. Kossoff, EH, and Rho, JM. Ketogenic diets: evidence from Short- and long-term efficacy. Neurotherapeutics. (2009) 6:406–14. doi: 10.1016/j.nurt.2009.01.005

PubMed Abstract | Crossref Full Text | Google Scholar

2. Taub, KS, Kessler, SK, and Bergqvist, AGC. Risk of seizure recurrence after achieving initial seizure freedom on the ketogenic diet. Epilepsia. (2014) 55:579–83. doi: 10.1111/epi.12583

PubMed Abstract | Crossref Full Text | Google Scholar

3. Caraballo, R, Vaccarezza, M, Cersósimo, R, Rios, V, Soraru, A, Arroyo, H, et al. Long-term follow-up of the ketogenic diet for refractory epilepsy: multicenter Argentinean experience in 216 pediatric patients. Seizure. (2011) 20:640–5. doi: 10.1016/j.seizure.2011.06.009

PubMed Abstract | Crossref Full Text | Google Scholar

4. Martinez, CC, Pyzik, PL, and Kossoff, EH. Discontinuing the ketogenic diet in seizure-free children: recurrence and risk factors. Epilepsia. (2007) 48:187–90. doi: 10.1111/j.1528-1167.2006.00911.x

PubMed Abstract | Crossref Full Text | Google Scholar

5. Patel, A, Pyzik, PL, Turner, Z, Rubenstein, JE, and Kossoff, EH. Long-term outcomes of children treated with the ketogenic diet in the past. Epilepsia. (2010) 51:1277–82. doi: 10.1111/j.1528-1167.2009.02488.x

PubMed Abstract | Crossref Full Text | Google Scholar

6. Schoeler, NE, Ridout, D, Neal, EG, Becirovic, M, Whiteley, VJ, Meskell, R, et al. Maintenance of response to ketogenic diet therapy for drug-resistant epilepsy post diet discontinuation: a multi-Centre case note review. Seizure. (2024) 121:78–84. doi: 10.1016/j.seizure.2024.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

7. Lusardi, TA, Akula, KK, Coffman, SQ, Ruskin, DN, Masino, SA, and Boison, D. Ketogenic diet prevents Epileptogenesis and disease progression in adult mice and rats. Neuropharmacology. (2015) 99:500–9. doi: 10.1016/j.neuropharm.2015.08.007

PubMed Abstract | Crossref Full Text | Google Scholar

8. Pawlik, MJ, Miziak, B, Walczak, A, Konarzewska, A, Chrościńska-Krawczyk, M, Albrecht, J, et al. Selected molecular targets for Antiepileptogenesis. Int J Mol Sci. (2021) 22:9737. doi: 10.3390/ijms22189737

PubMed Abstract | Crossref Full Text | Google Scholar

9. Neal, EG, Chaffe, H, Schwartz, RH, Lawson, MS, Edwards, N, Fitzsimmons, G, et al. The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial. Lancet Neurol. (2008) 7:500–6. doi: 10.1016/S1474-4422(08)70092-9

PubMed Abstract | Crossref Full Text | Google Scholar

10. Pavón, S, Lázaro, E, Martínez, O, Amayra, I, López-Paz, J, Caballero, P, et al. Ketogenic diet and cognition in neurological diseases: a systematic review. Nutr Rev. (2021) 79:802–13. doi: 10.1093/nutrit/nuaa113

PubMed Abstract | Crossref Full Text | Google Scholar

11. Halyburton, AK, Brinkworth, GD, Wilson, CJ, Noakes, M, Buckley, JD, Keogh, JB, et al. Low- and high-carbohydrate weight-loss diets have similar effects on mood but not cognitive performance. Am J Clin Nutr. (2007) 86:580–7. doi: 10.1093/ajcn/86.3.580

PubMed Abstract | Crossref Full Text | Google Scholar

12. Murphy, P, Likhodii, S, Nylen, K, and Burnham, WM. The antidepressant properties of the ketogenic diet. Biol Psychiatry. (2004) 56:981–3. doi: 10.1016/j.biopsych.2004.09.019

PubMed Abstract | Crossref Full Text | Google Scholar

13. Ijff, DF, Postulart, D, Lambrechts, DAJE, Majoie, MHJM, De Kinderen, RJA, Hendriksen, JGM, et al. Cognitive and behavioral impact of the ketogenic diet in children and adolescents with refractory epilepsy: a randomized controlled trial. Epilepsy Behav. (2016) 60:153–7. doi: 10.1016/j.yebeh.2016.04.033

PubMed Abstract | Crossref Full Text | Google Scholar

14. Davidson, TL, Hargrave, SL, Swithers, SE, Sample, CH, Fu, X, Kinzing, KP, et al. Inter-relationships among diet, obesity and hippocampal-dependent cognitive function. Neuroscience. (2013) 253:110–22. doi: 10.1016/j.neuroscience.2013.08.044

PubMed Abstract | Crossref Full Text | Google Scholar

15. Hallböök, T, Ji, S, Maudsley, S, and Martin, B. The effects of the ketogenic diet on behavior and cognition. Epilepsy Res. (2012) 100:304–9. doi: 10.1016/j.eplepsyres.2011.04.017

PubMed Abstract | Crossref Full Text | Google Scholar

16. Jiang, Y, Lu, Y, Jia, M, Wang, X, Zhang, Z, Hou, Q, et al. Ketogenic diet attenuates spatial and item memory impairment in Pentylenetetrazol-kindled rats. Brain Res. (2016) 1646:451–8. doi: 10.1016/j.brainres.2016.06.029

PubMed Abstract | Crossref Full Text | Google Scholar

17. Krikorian, R, Shidler, MD, Dangelo, K, Couch, SC, Benoit, SC, and Clegg, DJ. Dietary ketosis enhances memory in mild cognitive impairment. Neurobiol Aging. (2012) 33:425.e19–27. doi: 10.1016/j.neurobiolaging.2010.10.006

PubMed Abstract | Crossref Full Text | Google Scholar

18. Pan, Y, Larson, B, Araujo, JA, Lau, W, de Rivera, C, Santana, R, et al. Dietary supplementation with medium-chain TAG has long-lasting cognition-enhancing effects in aged dogs. Br J Nutr. (2010) 103:1746–54. doi: 10.1017/S0007114510000097

PubMed Abstract | Crossref Full Text | Google Scholar

19. Xu, K, Sun, X, Eroku, BO, Tsipis, CP, Puchowicz, MA, and LaManna, JC. Diet-induced ketosis improves cognitive performance in aged rats. Adv Exp Biol Med. (2010) 662:71–5. doi: 10.1007/978-1-4419-1241-1_9

PubMed Abstract | Crossref Full Text | Google Scholar

20. Ruskin, DN, Fortin, JA, Bisnauth, S, and Masino, SA. Ketogenic diets improve behaviors associated with autism Spectrum disorder in a sex-specific manner in the EL mouse. Physiol Behav. (2017) 168:138–45. doi: 10.1016/j.physbeh.2016.10.023

PubMed Abstract | Crossref Full Text | Google Scholar

21. Ruskin, DN, Murphy, MI, Slade, SL, and Masino, SA. Ketogenic diet improves behaviors in a maternal immune activation model of autism Spectrum disorder. PLoS One. (2017) 12:e0171643. doi: 10.1371/journal.pone.0171643

PubMed Abstract | Crossref Full Text | Google Scholar

22. Ruskin, DN, Svedova, J, Cote, JL, Sandau, U, Rho, JM, Kawamura, M, et al. Ketogenic diet improves Core symptoms of autism in BTBR mice. PLoS One. (2013) 8:e65021. doi: 10.1371/journal.pone.0065021

PubMed Abstract | Crossref Full Text | Google Scholar

23. Brady, M, Beltramini, A, Vaughan, G, and Bechard, AR. Benefits of a ketogenic diet on repetitive motor behavior in mice. Behav Brain Res. (2022) 422:113748. doi: 10.1016/j.bbr.2022.113748

PubMed Abstract | Crossref Full Text | Google Scholar

24. Ruskin, DN, Kawamura, M Jr, and Masino, SA. Reduced pain and inflammation in juvenile and adult rats fed a ketogenic diet. PLoS One. (2009) 4:e8349. doi: 10.1371/journal.pone.0008349

PubMed Abstract | Crossref Full Text | Google Scholar

25. Ruskin, DN, Suter, TACS, Ross, JL, and Masino, SA. Ketogenic diets and thermal pain: dissociation of Hypoalgesia, elevated ketones, and lowered glucose in rats. J Pain. (2013) 14:467–74. doi: 10.1016/j.jpain.2012.12.015

PubMed Abstract | Crossref Full Text | Google Scholar

26. Klejc, K, Cruz-Almeida, Y, and Sheffler, JL. Addressing pain using a Mediterranean ketogenic nutrition program in older adults with mild cognitive impairment. J Pain Res. (2024) 17:1867–80. doi: 10.2147/JPR.S451236

Crossref Full Text | Google Scholar

27. Elman, I, and Borsook, D. Common brain mechanisms of chronic pain and addiction. Neuron. (2016) 89:11–36. doi: 10.1016/j.neuron.2015.11.027

Crossref Full Text | Google Scholar

28. Sirven, J, Whedon, B, Caplan, D, Liporace, J, Glosser, D, O'Dwyer, J, et al. The ketogenic diet for intractable epilepsy in adults: preliminary results. Epilepsia. (1999) 40:1721–6. doi: 10.1111/j.1528-1157.1999.tb01589.x

PubMed Abstract | Crossref Full Text | Google Scholar

29. Lambrechts, DAJE, Bovens, MJM, de la Parra, NM, Hendriksen, JGM, Aldenkamp, AP, and Majoie, MJM. Ketogenic diet effects on cognition, mood, and psychosocial adjustment in children. Acta Neurol Scand. (2013) 127:103–8. doi: 10.1111/j.1600-0404.2012.01686.x

PubMed Abstract | Crossref Full Text | Google Scholar

30. Remahl, S, Dahlin, MG, and Åmark, PE. Influence of the ketogenic diet on 24-hour electroencephalogram in children with epilepsy. Pediatr Neurol. (2008) 38:38–43. doi: 10.1016/j.pediatrneurol.2007.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

31. Pulsifer, MB, Gordon, JM, Brandt, J, Vining, EPG, and Freeman, JM. Effects of ketogenic diet on development and behavior: preliminary report of a prospective study. Dev Med Child Neurol. (2001) 43:301–6. doi: 10.1111/j.1469-8749.2001.tb00209.x

PubMed Abstract | Crossref Full Text | Google Scholar

32. Masino, SA, Ruskin, DN, Freedgood, NR, Lindefeldt, M, and Dahlin, M. Differential ketogenic diet-induced shift in CSF lipid/carbohydrate metabolome of pediatric epilepsy patients with optimal vs. no anticonvulsant response: a pilot study. Nutr Metab. (2021) 18:23. doi: 10.1186/s12986-020-00524-1

PubMed Abstract | Crossref Full Text | Google Scholar

33. Lloyd, HGE, Lindström, K, and Fredholm, BB. Intracellular formation and release of adenosine from rat hippocampal slices evoked by electrical stimulation or energy depletion. Neurochem Int. (1993) 23:173–85. doi: 10.1016/0197-0186(93)90095-m

PubMed Abstract | Crossref Full Text | Google Scholar

34. Dulla, CG, Frenguelli, BG, Staley, KJ, and Masino, SA. Intracellular acidification causes adenosine release during states of Hyperexcitability in the Hippocampus. J Neurophysiol. (2009) 102:1984–93. doi: 10.1152/jn.90695.2008

PubMed Abstract | Crossref Full Text | Google Scholar

35. Masino, SA, Kawamura, M Jr, Wasser, CD, Pomeroy, LT, and Ruskin, DN. Adenosine, ketogenic diet and epilepsy: the emerging therapeutic relationship between metabolism and brain activity. Curr Neuropharmacol. (2009) 7:257–68. doi: 10.2174/157015909789152164

PubMed Abstract | Crossref Full Text | Google Scholar

36. Masino, SA, and Geiger, JD. Are purines mediators of the anticonvulsant/neuroprotective effects of ketogenic diets? Trends Neurosci. (2008) 31:273–8. doi: 10.1016/j.tins.2008.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

37. Kawamura, M Jr, Ruskin, DN, Geiger, JD, Boison, D, and Masino, SA. Ketogenic diet sensitizes glucose control of hippocampal excitability. J Lipid Res. (2014) 55:2254–60. doi: 10.1194/jlr.M046755

PubMed Abstract | Crossref Full Text | Google Scholar

38. Masino, SA, Li, T, Theofilas, P, Sandau, U, Ruskin, DN, Fredholm, BB, et al. A ketogenic diet suppresses seizures in mice through adenosine A1 receptors. J Clin Invest. (2011) 121:2679–83. doi: 10.1172/JCI57813

PubMed Abstract | Crossref Full Text | Google Scholar

39. Kawamura, M Jr, Ruskin, DN, and Masino, SA. Metabolic autocrine regulation of neurons involves cooperation among Pannexin Hemichannels, adenosine receptors and KATP channels. J Neurosci. (2010) 30:3886–95. doi: 10.1523/JNEUROSCI.0055-10.2010

PubMed Abstract | Crossref Full Text | Google Scholar

40. Socała, K, Nieoczym, D, Pieróg, M, and Wlaź, P. Role of the adenosine system and glucose restriction in the acute anticonvulsant effect of Caprylic acid in the 6hz psychomotor seizure test in mice. Prog Neuro-Psychopharmacol Biol Psychiatry. (2015) 57:44–51. doi: 10.1016/j.pnpbp.2014.10.006

PubMed Abstract | Crossref Full Text | Google Scholar

41. Kovács, Z, D'Agostino, DP, Dobolyi, A, and Ari, C. Adenosine A1 receptor antagonism abolished the anti-seizure effects of exogenous ketone supplementation in Wistar albino Glaxo Rijswijk rats. Front Mol Neurosci. (2017) 10:235. doi: 10.3389/fnmol.2017.00235

PubMed Abstract | Crossref Full Text | Google Scholar

42. Yang, Q, Guo, M, Wang, X, Zhao, Y, Zhao, Q, Ding, H, et al. Ischemic preconditioning with a ketogenic diet improves brain ischemic tolerance through increased extracellular adenosine levels and hypoxia-inducible factors. Brain Res. (2017) 1667:11–8. doi: 10.1016/j.brainres.2017.04.010

PubMed Abstract | Crossref Full Text | Google Scholar

43. Deroche-Gamonet, V, Belin, D, and Piazza, PV. Evidence for addiction-like behavior in the rat. Science. (2004) 305:1014–7. doi: 10.1126/science.1099020

PubMed Abstract | Crossref Full Text | Google Scholar

44. Vanderschuren, LJMJ, and Everitt, BJ. Drug seeking becomes compulsive after prolonged cocaine Self-administration. Science. (2004) 305:1017–9. doi: 10.1126/science.1098975

PubMed Abstract | Crossref Full Text | Google Scholar

45. Grimm, JW, Hope, BT, Wise, RA, and Shaham, Y. Incubation of cocaine craving after withdrawal. Nature. (2001) 412:141–2. doi: 10.1038/35084134

PubMed Abstract | Crossref Full Text | Google Scholar

46. Volkow, ND, Fowler, JS, and Wang, G-J. Imaging studies on the role of dopamine in cocaine reinforcement and addiction in humans. J Psychopharmacol. (1999) 13:337–45. doi: 10.1177/026988119901300406

PubMed Abstract | Crossref Full Text | Google Scholar

47. Porrino, LJ, Lyons, D, Smith, HR, Daunais, JB, and Nader, MA. Cocaine Self-administration produces a progressive involvement of limbic, association, and sensorimotor striatal domains. J Neurosci. (2004) 24:3554–62. doi: 10.1523/JNEUROSCI.5578-03.2004

PubMed Abstract | Crossref Full Text | Google Scholar

48. Lu, L, Grimm, JW, Hope, BT, and Shaham, Y. Incubation of cocaine craving after withdrawal: a review of preclinical data. Neuropharmacology. (2004) 47:214–26. doi: 10.1016/j.neuropharm.2004.06.027

PubMed Abstract | Crossref Full Text | Google Scholar

49. Franco, R, and Navarro, G. Adenosine A2A receptor antagonists in neurodegenerative diseases: huge potential and huge challenges. Front Psych. (2018) 9:68. doi: 10.3389/fpsyt.2018.00068

PubMed Abstract | Crossref Full Text | Google Scholar

50. Rivera-Oliver, M, and Díaz-Ríos, M. Using caffeine and other adenosine receptor antagonists and agonists as therapeutic tools against neurodegenerative diseases: a review. Life Sci. (2014) 101:1–9. doi: 10.1016/j.lfs.2014.01.083

PubMed Abstract | Crossref Full Text | Google Scholar

51. Ferré, S, O'Connor, WT, Svenningsson, P, Björklund, L, Lindberg, J, Tinner, B, et al. Dopamine D1 receptor-mediated facilitation of GABAergic neurotransmission in the rat Strioentopeduncular pathway and its modulation by adenosine A1 receptor-mediated mechanisms. Eur J Neurosci. (1996) 8:1545–53. doi: 10.1111/j.1460-9568.1996.tb01617.x

PubMed Abstract | Crossref Full Text | Google Scholar

52. Fink, JS, Weaver, DR, Rivkees, SA, Peterfreund, RA, Pollack, AE, Adler, EM, et al. Molecular cloning of the rat A2 adenosine receptor: selective co-expression with D2 dopamine receptors in rat striatum. Mol Brain Res. (1992) 14:186–95. doi: 10.1016/0169-328x(92)90173-9

PubMed Abstract | Crossref Full Text | Google Scholar

53. Franco, R, Ferré, S, Agnati, L, Torvinen, M, Ginés, S, Hillion, J, et al. Evidence for adenosine/dopamine receptor interactions: indications for Heteromerization. Neuropsychopharmacology. (2000) 23:S50–9. doi: 10.1016/S0893-133X(00)00144-5

PubMed Abstract | Crossref Full Text | Google Scholar

54. Ginés, S, Hillion, J, Torvinen, M, Crom, SL, Casado, V, Canela, EI, et al. Dopamine D1 and adenosine A1 receptors form functionally interacting Heteromeric complexes. Proc Natl Acad Sci USA. (2000) 97:8606–11. doi: 10.1073/pnas.150241097

PubMed Abstract | Crossref Full Text | Google Scholar

55. Hillion, J, Canals, M, Torvinen, M, Casadó, V, Scott, R, Terasmaa, A, et al. Coaggregation, Cointernalization, and Codesensitization of adenosine A2A receptors and dopamine D2 receptors. J Biol Chem. (2002) 277:18091–7. doi: 10.1074/jbc.M107731200

PubMed Abstract | Crossref Full Text | Google Scholar

56. Prasad, K, de Vries, EFJ, Elsinga, PH, Dierckx, R, and van Waarde, A. Allosteric interactions between adenosine A2Aa and dopamine D2 receptors in Heteromeric complexes: biochemical and pharmacological characteristics, and opportunities for PET imaging. Int J Mol Sci. (2021) 22:1719. doi: 10.3390/ijms22041719

PubMed Abstract | Crossref Full Text | Google Scholar

57. Filip, M, Frankowska, M, Zaniewska, M, Przegaliński, E, Műller, C, Agnati, L, et al. Involvement of adenosine A2A and dopamine receptors in the locomotor and sensitizing effects of cocaine. Brain Res. (2006) 1077:67–80. doi: 10.1016/j.brainres.2006.01.038

PubMed Abstract | Crossref Full Text | Google Scholar

58. Shen, H-Y, Coelho, JE, Ohtsuka, N, Canas, PM, Day, Y-J, Huang, Q-Y, et al. A critical role of the adenosine A2A receptor in Extrastriatal neurons in modulating psychomotor activity as revealed by opposite phenotypes of striatum and forebrain A2A receptor Knock-outs. J Neurosci. (2008) 28:2970–5. doi: 10.1523/JNEUROSCI.5255-07.2008

PubMed Abstract | Crossref Full Text | Google Scholar

59. O'Neill, CE, Levis, SC, Schreiner, DC, Amat, J, Maier, SF, and Bachtell, RK. Effects of adolescent caffeine consumption on cocaine sensitivity. Neuropsychopharmacology. (2015) 40:813–21. doi: 10.1038/npp.2014.278

PubMed Abstract | Crossref Full Text | Google Scholar

60. Acton, D, Broadhead, MJ, and Miles, GB. Modulation of spinal motor networks by astrocyte-derived adenosine is dependent on D1-like dopamine receptor signaling. J Neurophysiol. (2018) 120:998–1009. doi: 10.1152/jn.00783.2017

PubMed Abstract | Crossref Full Text | Google Scholar

61. Muñiz, J, Gomez, G, González, B, Rivero-Echeto, MC, Cadet, JL, García-Rill, E, et al. Combined effects of simultaneous exposure to caffeine and cocaine in the mouse striatum. Neurotox Res. (2016) 29:525–38. doi: 10.1007/s12640-016-9601-0

PubMed Abstract | Crossref Full Text | Google Scholar

62. Hobson, BD, Merritt, KE, and Bachtell, RK. Stimulation of adenosine receptors in the nucleus Accumbens reverses the expression of cocaine sensitization and Cross-sensitization to dopamine D2 receptors in rats. Neuropharmacology. (2012) 63:1172–81. doi: 10.1016/j.neuropharm.2012.06.038

PubMed Abstract | Crossref Full Text | Google Scholar

63. Wydra, K, Golembiowska, K, Suder, A, Kaminska, K, Fuxe, K, and Filip, M. On the role of adenosine (a)2A receptors in cocaine-induced reward: a pharmacological and neurochemical analysis in rats. Psychopharmacology. (2015) 232:421–35. doi: 10.1007/s00213-014-3675-2

PubMed Abstract | Crossref Full Text | Google Scholar

64. Hobson, BD, O'Neill, CE, Levis, SC, Monteggia, LM, Neve, RL, Self, DW, et al. Adenosine A1 and dopamine D1 receptor regulation of AMPA receptor phosphorylation and cocaine-seeking behavior. Neuropsychopharmacology. (2013) 38:1974–83. doi: 10.1038/npp.2013.96

PubMed Abstract | Crossref Full Text | Google Scholar

65. Derlet, RW, Tseng, JC, and Albertson, TE. Potentiation of cocaine and d-amphetamine toxicity with caffeine. Am J Emerg Med. (1992) 10:211–6. doi: 10.1016/0735-6757(92)90211-F

PubMed Abstract | Crossref Full Text | Google Scholar

66. Salamone, JD, Correa, M, Ferrigno, S, Yang, JH, Rotolo, RA, and Presby, RE. The psychopharmacology of effort-related decision making: dopamine, adenosine, and insights into the neurochemistry of motivation. Pharmacol Rev. (2018) 70:747–62. doi: 10.1124/pr.117.015107

PubMed Abstract | Crossref Full Text | Google Scholar

67. Dahlin, M, Månsson, J-E, and Åmark, P. CSF levels of dopamine and serotonin, but not norepinephrine, metabolites are influenced by the ketogenic diet in children with epilepsy. Epilepsy Res. (2012) 99:132–8. doi: 10.1016/j.eplepsyres.2011.11.003

PubMed Abstract | Crossref Full Text | Google Scholar

68. Church, WH, Adams, RE, and Wyss, LS. Ketogenic diet alters dopaminergic activity in the mouse cortex. Neurosci Lett. (2014) 571:1–4. doi: 10.1016/j.neulet.2014.04.016

PubMed Abstract | Crossref Full Text | Google Scholar

69. Volkow, ND, and Morales, M. The brain on drugs: from reward to addiction. Cell. (2015) 162:712–25. doi: 10.1016/j.cell.2015.07.046

PubMed Abstract | Crossref Full Text | Google Scholar

70. Baik, JH. Dopamine signaling in reward-related behaviors. Front Neural Circuits. (2013) 7:152. doi: 10.3389/fncir.2013.00152

PubMed Abstract | Crossref Full Text | Google Scholar

71. Morava, E, Gardeitchik, T, Kozicz, T, de Boer, L, Koene, S, de Vries, MC, et al. Depressive behaviour in children diagnosed with a mitochondrial disorder. Mitochondrion. (2010) 10:528–33. doi: 10.1016/j.mito.2010.05.011

PubMed Abstract | Crossref Full Text | Google Scholar

72. Meles, SK, Teune, LK, de Jong, BM, Dierckx, RA, and Leenders, KL. Metabolic imaging in Parkinson disease. J Nucl Med. (2017) 58:23–8. doi: 10.2967/jnumed.116.183152

PubMed Abstract | Crossref Full Text | Google Scholar

73. Talbot, K, Wang, HY, Kazi, H, Han, LY, Bakshi, KP, Stucky, A, et al. Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. (2012) 122:1316–38. doi: 10.1172/JCI59903

PubMed Abstract | Crossref Full Text | Google Scholar

74. Guest, PC, Schwarz, E, Krishnamurthy, D, Harris, LW, Leweke, FM, Rothermundt, M, et al. Altered levels of circulating insulin and other neuroendocrine hormones associated with the onset of schizophrenia. Psychoneuroendocrinology. (2011) 36:1092–6. doi: 10.1016/j.psyneuen.2010.12.018

PubMed Abstract | Crossref Full Text | Google Scholar

75. Chauhan, A, Gu, F, Essa, MM, Wegiel, J, Kaur, K, Brown, WT, et al. Brain region-specific deficit in mitochondrial Electron transport chain complexes in children with autism. J Neurochem. (2011) 117:209–20. doi: 10.1111/j.1471-4159.2011.07189.x

PubMed Abstract | Crossref Full Text | Google Scholar

76. 8th Global Symposium on Ketogenic Therapies. San Diego, California, USA: International Neurological Ketogenic Society. (2023).

Google Scholar

77. Newport, MT, VanItallie, TB, King, MT, and Veech, RL. A new way to produce hyperketonemia: use of ketone Ester in a case of Alzheimer’s disease. Alzheimers Dement. (2015) 11:99–103. doi: 10.1016/j.jalz.2014.01.006

PubMed Abstract | Crossref Full Text | Google Scholar

78. Morrill, SJ, and Gibas, KJ. Ketogenic diet rescues cognition in ApoE4+ patient with mild Alzheimer's disease: a case study. Diabetes Metab Syndr. (2019) 13:1187–91. doi: 10.1016/j.dsx.2019.01.035

PubMed Abstract | Crossref Full Text | Google Scholar

79. Tai, J, Liu, W, Li, Y, Li, L, and Holscher, C. Neuroprotective effects of a triple GLP-1/GIP/glucagon receptor agonist in the APP/PS1 transgenic mouse model of Alzheimer's disease. Brain Res. (2018) 1678:64–74. doi: 10.1016/j.brainres.2017.10.012

PubMed Abstract | Crossref Full Text | Google Scholar

80. Combs, CK, Johnson, DE, Karlo, JC, Cannady, SB, and Landreth, GE. Inflammatory mechanisms in Alzheimer's disease: inhibition of β-amyloid-stimulated Proinflammatory responses and neurotoxicity by PPARγ agonists. J Neurosci. (2000) 20:558–67. doi: 10.1523/JNEUROSCI.20-02-00558.2000

PubMed Abstract | Crossref Full Text | Google Scholar

81. Lindberg, D, Shan, D, Ayers-Ringler, J, Oliveros, A, Benitez, J, Prieto, M, et al. Purinergic signaling and energy homeostasis in psychiatric disorders. Curr Mol Med. (2015) 15:275–95. doi: 10.2174/1566524015666150330163724

PubMed Abstract | Crossref Full Text | Google Scholar

82. Cunnane, SC, Trushina, E, Morland, C, Prigione, A, Casadesus, G, Andrews, ZB, et al. Brain energy rescue: an emerging therapeutic concept for neurodegenerative disorders of ageing. Nat Rev Drug Discov. (2020) 19:609–33. doi: 10.1038/s41573-020-0072-x

PubMed Abstract | Crossref Full Text | Google Scholar

83. DeVivo, DC, Leckie, MP, Ferrendelli, JS, and McDougal, DB Jr. Chronic ketosis and cerebral metabolism. Ann Neurol. (1978) 3:331–7. doi: 10.1002/ana.410030410

PubMed Abstract | Crossref Full Text | Google Scholar

84. Nakazawa, M, Kodama, S, and Matsuo, T. Effects of ketogenic diet on electroconvulsive threshold and brain contents of adenosine nucleotides. Brain and Development. (1983) 5:375–80. doi: 10.1016/s0387-7604(83)80042-4

PubMed Abstract | Crossref Full Text | Google Scholar

85. Deng-Bryant, Y, Prins, ML, Hovda, DA, and Harris, NG. Ketogenic diet prevents alterations in brain metabolism in young but not adult rats after traumatic brain injury. J Neurotrauma. (2011) 28:1813–25. doi: 10.1089/neu.2011.1822

PubMed Abstract | Crossref Full Text | Google Scholar

86. Nylen, K, Velazquez, JLP, Sayed, V, Gibson, KM, Burnham, WM, and Snead, OC III. The effects of a ketogenic diet on ATP concentrations and the number of hippocampal mitochondria in Aldh5a1−/− mice. Biochim Biophys Acta. (2009) 1790:208–12. doi: 10.1016/j.bbagen.2008.12.005

PubMed Abstract | Crossref Full Text | Google Scholar

87. Zhao, Z, Lange, DJ, Voustianiouk, A, MacGrogan, D, Ho, L, Suh, J, et al. A ketogenic diet as a potential novel therapeutic intervention in amyotrophic lateral sclerosis. BMC Neurosci. (2006) 7:29. doi: 10.1186/1471-2202-7-29

PubMed Abstract | Crossref Full Text | Google Scholar

88. Miller, VJ, LaFountain, RA, Barnhart, E, Sapper, TS, Short, J, Arnold, WD, et al. A ketogenic diet combined with exercise alters mitochondrial function in human skeletal muscle while improving metabolic health. Am J Physiol Endocrinol Metab. (2020) 319:E995–E1007. doi: 10.1152/ajpendo.00305.2020

PubMed Abstract | Crossref Full Text | Google Scholar

89. Bough, KJ, Wetherington, J, Hassel, B, Pare, JF, Gawryluk, JW, Greene, JG, et al. Mitochondrial biogenesis in the anticonvulsant mechanism of the ketogenic diet. Ann Neurol. (2006) 60:223–35. doi: 10.1002/ana.20899

PubMed Abstract | Crossref Full Text | Google Scholar

90. Noh, HS, Lee, HP, Kim, DW, Kang, SS, Cho, GJ, Rho, JM, et al. A cDNA microarray analysis of gene expression profiles in rat Hippocampus following a ketogenic diet. Mol Brain Res. (2004) 129:80–7. doi: 10.1016/j.molbrainres.2004.06.020

PubMed Abstract | Crossref Full Text | Google Scholar

91. Ahn, Y, Narous, M, Tobias, R, Rho, JM, and Mychasiuk, R. The ketogenic diet modifies social and metabolic alterations identified in the prenatal Valproic acid model of autism Spectrum disorder. Dev Neurosci. (2014) 36:371–80. doi: 10.1159/000362645

PubMed Abstract | Crossref Full Text | Google Scholar

92. Ahola-Erkkila, S, Carroll, C, Peltola-Mjosund, K, Tulkki, V, Mattila, I, Seppanen-Laasko, T, et al. Ketogenic diet slows down mitochondrial myopathy progression in mice. Hum Mol Genet. (2010) 19:1974–84. doi: 10.1093/hmg/ddq076

PubMed Abstract | Crossref Full Text | Google Scholar

93. Balietti, M, Giorgetti, B, di, G, Casoli, T, Platano, D, Solazzi, M, et al. A ketogenic diet increases succinic dehydrogenase (SDH) activity and recovers age-related decrease in numeric density of SDH-positive mitochondria in cerebellar Purkinje cells of late-adult rats. Micron. (2010) 41:143–8. doi: 10.1016/j.micron.2009.08.010

PubMed Abstract | Crossref Full Text | Google Scholar

94. Sullivan, PG, Rippy, NA, Dorenbos, K, Concepcion, RC, Agarwal, AK, and Rho, JM. The ketogenic diet increases mitochondrial uncoupling protein levels and activity. Ann Neurol. (2004) 55:576–80. doi: 10.1002/ana.20062

PubMed Abstract | Crossref Full Text | Google Scholar

95. Hasan-Olive, MM, Lauritzen, KH, Ali, M, Rasmussen, LJ, Storm-Mathisen, J, and Bergersen, LH. A ketogenic diet improves mitochondrial biogenesis and bioenergetics via the PGC1α-SIRT3-UCP2 Axis. Neurochem Res. (2019) 44:22–37. doi: 10.1007/s11064-018-2588-6

PubMed Abstract | Crossref Full Text | Google Scholar

96. Seira, O, Kolehmainin, K, Liu, J, Streijger, F, Haegart, A, Lebihan, S, et al. Ketogenesis controls mitochondrial gene expression and rescues mitochondrial bioenergetics after cervical spinal cord injury in rats. Sci Rep. (2021) 11:16359. doi: 10.1038/s41598-021-96003-5

PubMed Abstract | Crossref Full Text | Google Scholar

97. Wang, B-H, Hou, Q, Lu, Y-Q, Jia, M-M, Qiu, T, Wang, X-H, et al. Ketogenic diet attenuates neuronal injury via autophagy and mitochondrial pathways in Pentylenetetrazol-kindled seizures. Brain Res. (2018) 1678:106–15. doi: 10.1016/j.brainres.2017.10.009

PubMed Abstract | Crossref Full Text | Google Scholar

98. Kim, H, Kim, YK, Lee, JY, Choi, AR, Kim, DJ, and Choi, JS. Hypometabolism and altered metabolic connectivity in patients with internet gaming disorder and alcohol use disorder. Prog Neuro-Psychopharmacol Biol Psychiatry. (2019) 95:109680. doi: 10.1016/j.pnpbp.2019.109680

PubMed Abstract | Crossref Full Text | Google Scholar

99. Volkow, ND, Hitzemann, R, Wang, GJ, Fowler, JS, Wolf, AP, Dewey, SL, et al. Long-term frontal brain metabolic changes in cocaine abusers. Synapse. (1992) 11:184–90. doi: 10.1002/syn.890110303

Crossref Full Text | Google Scholar

100. Chang, L, Alicata, D, Ernst, T, and Volkow, N. Structural and metabolic brain changes in the striatum associated with methamphetamine abuse. Addiction. (2007) 102:16–32. doi: 10.1111/j.1360-0443.2006.01782.x

PubMed Abstract | Crossref Full Text | Google Scholar

101. Wabik, A, Trypka, E, Bladowska, J, Statkiewicz, M, Sasiadek, M, and Zimny, A. Comparison of dynamic susceptibility contrast enhanced MR and FDG-PET brain studies in patients with Alzheimer's disease and amnestic mild cognitive impairment. J Transl Med. (2022) 20:259. doi: 10.1186/s12967-022-03464-x

PubMed Abstract | Crossref Full Text | Google Scholar

102. Anderson, ND. State of the science on mild cognitive impairment (MCI). CNS Spectr. (2019) 24:78–87. doi: 10.1017/S1092852918001347

PubMed Abstract | Crossref Full Text | Google Scholar

103. Bi, Q, Wang, W, Niu, N, Li, H, Wang, Y, Huang, W, et al. Relationship between the disrupted topological efficiency of the structural brain connectome and glucose Hypometabolism in Normal aging. NeuroImage. (2021) 226:117591. doi: 10.1016/j.neuroimage.2020.117591

PubMed Abstract | Crossref Full Text | Google Scholar

104. Noakes, M, Foster, PR, Keogh, JB, James, AP, Mamo, JC, and Clifton, PM. Comparison of Isocaloric very low carbohydrate/high saturated fat and high carbohydrate/low saturated fat diets on body composition and cardiovascular risk. Nutr Metab. (2006) 3:7. doi: 10.1186/1743-7075-3-7

PubMed Abstract | Crossref Full Text | Google Scholar

105. Nuttall, FQ, Almokayyad, RM, and Gannon, MC. Comparison of a carbohydrate-free diet vs. fasting on plasma glucose, insulin and glucagon in type 2 diabetes. Metabolism. (2015) 64:253–62. doi: 10.1016/j.metabol.2014.10.004

PubMed Abstract | Crossref Full Text | Google Scholar

106. Tay, J, Luscombe-Marsh, ND, Thompson, CH, Noakes, M, Buckley, JD, Wittert, GA, et al. Comparison of low- and high-carbohydrate diets for type 2 diabetes management: a randomized trial. Am J Clin Nutr. (2015) 102:780–90. doi: 10.3945/ajcn.115.112581

PubMed Abstract | Crossref Full Text | Google Scholar

107. Nolan, J, Rush, A, and Kaye, J. Glycaemic stability of a cyclist with type 1 diabetes: 4011 km in 20 days on a ketogenic diet. Diabet Med. (2019) 36:1503–7. doi: 10.1111/dme.14049

PubMed Abstract | Crossref Full Text | Google Scholar

108. Saslow, LR, Daubenmier, JJ, Moskowitz, JT, Kim, S, Murphy, EJ, Phinney, SD, et al. Twelve-month outcomes of a randomized trial of a moderate-carbohydrate versus very low-carbohydrate diet in overweight adults with type 2 diabetes mellitus or prediabetes. Nutr Diabetes. (2017) 7:304. doi: 10.1038/s41387-017-0006-9

PubMed Abstract | Crossref Full Text | Google Scholar

109. Webster, CC, Murphy, TE, Larmuth, KM, Noakes, TD, and Smith, JA. Diet, diabetes status, and personal experiences of individuals with type 2 diabetes who Self-selected and followed a low carbohydrate high fat diet. Diabetes Metab Syndr Obes. (2019) 12:2567–82. doi: 10.2147/DMSO.S227090

PubMed Abstract | Crossref Full Text | Google Scholar

110. Kiyatkin, EA, and Lenoir, M. Rapid fluctuations in extracellular brain glucose levels induced by natural arousing stimuli and intravenous cocaine: fueling the brain during neural activation. J Neurophysiol. (2012) 108:1669–84. doi: 10.1152/jn.00521.2012

PubMed Abstract | Crossref Full Text | Google Scholar

111. de Carvalho Vidigal, F, Cocate, PG, Pereira, LG, and Alfenas, CG. The role of hyperglycemia in the induction of oxidative stress and inflammatory process. Nutr Hosp. (2012) 27:1391–8. doi: 10.3305/nh.2012.27.5.5917

PubMed Abstract | Crossref Full Text | Google Scholar

112. Nedosugova, LV, Markina, YV, Bochkareva, LA, Kuzina, IA, Petunina, NA, Yudina, IY, et al. Inflammatory mechanisms of diabetes and its vascular complications. Biomedicines. (2022) 10:1168. doi: 10.3390/biomedicines10051168

PubMed Abstract | Crossref Full Text | Google Scholar

113. Martin, C, Tansey, KE, Schalkwyk, LC, and Powell, TR. The inflammatory cytokines: molecular biomarkers for major depressive disorder? Biomark Med. (2015) 9:169–80. doi: 10.2217/bmm.14.29

PubMed Abstract | Crossref Full Text | Google Scholar

114. Tendler, D, Lin, S, Yancy, WS Jr, Mavropoulos, J, Sylvestre, P, Rockey, DC, et al. The effect of a low-carbohydrate, ketogenic diet on nonalcoholic fatty liver disease: a pilot study. Dig Dis Sci. (2007) 52:589–93. doi: 10.1007/s10620-006-9433-5

PubMed Abstract | Crossref Full Text | Google Scholar

115. Pérez-Guisado, J, and Muñoz-Serrano, A. The effect of the Spanish ketogenic Mediterranean diet on nonalcoholic fatty liver disease: a pilot study. J Med Food. (2011) 14:677–80. doi: 10.1089/jmf.2011.0075

PubMed Abstract | Crossref Full Text | Google Scholar

116. Forsythe, CE, Phinney, SD, Fernandez, ML, Quann, EE, Wood, RJ, Bibus, DM, et al. Comparison of low fat and low carbohydrate diets on circulating fatty acid composition and markers of inflammation. Lipids. (2008) 43:65–77. doi: 10.1007/s11745-007-3132-7

Crossref Full Text | Google Scholar

117. Field, R, Pourkazemi, F, and Rooney, K. Effects of a low-carbohydrate ketogenic diet on reported pain, blood biomarkers and quality of life in patients with chronic pain: a pilot randomised clinical trial. Pain Med. (2022) 23:326–38. doi: 10.1093/pm/pnab278

PubMed Abstract | Crossref Full Text | Google Scholar

118. Ruskin, DN, and Masino, SA. The nervous system and metabolic dysregulation: emerging evidence converges on ketogenic diet therapy. Front Neurosci. (2012) 6:33. doi: 10.3389/fnins.2012.00033

PubMed Abstract | Crossref Full Text | Google Scholar

119. Nandivada, P, Fell, GL, Pan, AH, Nose, V, Ling, P-R, Bistrian, BR, et al. Eucaloric ketogenic diet reduces hypoglycemia and inflammation in mice with Endotoxemia. Lipids. (2016) 51:703–14. doi: 10.1007/s11745-016-4156-7

Crossref Full Text | Google Scholar

120. Ruskin, DN, Sturdevant, IC, Wyss, LS, and Masino, SA. Ketogenic diet effects on inflammatory allodynia and ongoing pain in rodents. Sci Rep. (2021) 11:725. doi: 10.1038/s41598-020-80727-x

PubMed Abstract | Crossref Full Text | Google Scholar

121. Kong, G, Wang, J, Li, R, Huang, Z, and Wang, L. Ketogenic diet ameliorates inflammation by inhibiting the NLRP3 Inflammasome in osteoarthritis. Arthritis Res Ther. (2022) 24:113. doi: 10.1186/s13075-022-02802-0

PubMed Abstract | Crossref Full Text | Google Scholar

122. Jeong, EA, Jeon, BT, Shin, HJ, Kim, N, Lee, DH, Kim, HJ, et al. Ketogenic diet-induced peroxisome proliferator-activated receptor-γ activation decreases Neuroinflammation in the mouse Hippocampus after Kainic acid-induced seizures. Exp Neurol. (2011) 232:195–202. doi: 10.1016/j.expneurol.2011.09.001

PubMed Abstract | Crossref Full Text | Google Scholar

123. Kim, DY, Hao, J, Liu, R, Turner, G, Shi, FD, and Rho, JM. Inflammation-mediated memory dysfunction and effects of a ketogenic diet in a murine model of multiple sclerosis. PLoS One. (2012) 7:e35476. doi: 10.1371/journal.pone.0035476

PubMed Abstract | Crossref Full Text | Google Scholar

124. Dilimulati, D, Zhang, F, Shao, S, Lv, T, Lu, Q, Cao, M, et al. Ketogenic diet modulates Neuroinflammation via metabolites from Lactobacillus reuteri after repetitive mild traumatic brain injury in adolescent mice. Cell Mol Neurobiol. (2022) 43:907–23. doi: 10.1007/s10571-022-01226-3

PubMed Abstract | Crossref Full Text | Google Scholar

125. Dupuis, N, Curatolo, N, Benoist, J-F, and Auvin, S. Ketogenic diet exhibits anti-inflammatory properties. Epilepsia. (2015) 56:e95–8. doi: 10.1111/epi.13038

PubMed Abstract | Crossref Full Text | Google Scholar

126. Jiang, Z, Yin, X, Wang, M, Chen, T, Wang, Y, Gao, Z, et al. Effects of ketogenic diet on Neuroinflammation in neurodegenerative diseases. Aging Dis. (2022) 13:1146–65. doi: 10.14336/AD.2021.1217

PubMed Abstract | Crossref Full Text | Google Scholar

127. Martinez, LA, Lees, ME, Ruskin, DN, and Masino, SA. The ketogenic diet decreases behavioral responses to cocaine in male and female rats. Neuropharmacology. (2019) 149:27–34. doi: 10.1016/j.neuropharm.2019.02.001

PubMed Abstract | Crossref Full Text | Google Scholar

128. Rodenas-Gonzalez, F, Blanco-Gandia, MC, Minarro, J, and Rodriguez-Arias, M. Effects of ketosis on cocaine-induced reinstatement in male mice. Neurosci Lett. (2022) 778:136619. doi: 10.1016/j.neulet.2022.136619

PubMed Abstract | Crossref Full Text | Google Scholar

129. Wiers, CE, Vendruscolo, LF, van der Veen, JW, Manza, P, Shokri-Kojori, E, Kroll, DS, et al. Ketogenic diet reduces alcohol withdrawal symptoms in humans and alcohol intake in rodents. Sci Adv. (2021) 7:eabf6780. doi: 10.1126/sciadv.abf6780

PubMed Abstract | Crossref Full Text | Google Scholar

130. Dencker, D, Molander, A, Thomsen, M, Schlumberger, C, Wortwein, G, Weikop, P, et al. Ketogenic diet suppresses alcohol withdrawal syndrome in rats. Alcohol Clin Exp Res. (2018) 42:270–7. doi: 10.1111/acer.13560

PubMed Abstract | Crossref Full Text | Google Scholar

131. Tonetto, S, Weikop, P, and Thomsen, M. Nutritional ketosis as treatment for alcohol withdrawal symptoms in female C57BL/6J mice. Sci Rep. (2024) 14:5092. doi: 10.1038/s41598-024-55310-3

PubMed Abstract | Crossref Full Text | Google Scholar

132. Bornebusch, AB, Mason, GF, Tonetto, S, Damsgaard, J, Gjedde, A, Fink-Jensen, A, et al. Effects of ketogenic diet and ketone monoester supplement on acute alcohol withdrawal symptoms in male mice. Psychopharmacology. (2021) 238:833–44. doi: 10.1007/s00213-020-05735-1

PubMed Abstract | Crossref Full Text | Google Scholar

133. Mahajan, VR, Elvig, SK, Vendruscolo, LF, Koob, GF, Darcey, VL, King, MT, et al. Nutritional ketosis as a potential treatment for alcohol use disorder. Front Psych. (2021) 12:781668. doi: 10.3389/fpsyt.2021.781668

PubMed Abstract | Crossref Full Text | Google Scholar

134. Nam, HW, Bruner, RC, and Choi, DS. Adenosine signaling in striatal circuits and alcohol use disorders. Mol Cells. (2013) 36:195–202. doi: 10.1007/s10059-013-0192-9

PubMed Abstract | Crossref Full Text | Google Scholar

135. Roberts, BM, Lambert, E, Livesey, JA, Wu, Z, Li, Y, and Cragg, SJ. Dopamine release in nucleus Accumbens is under tonic inhibition by adenosine A1 receptors regulated by astrocytic ENT1 and dysregulated by ethanol. J Neurosci. (2022) 42:1738–51. doi: 10.1523/JNEUROSCI.1548-21.2021

PubMed Abstract | Crossref Full Text | Google Scholar

136. Wu, M, Sahbaie, P, Zheng, M, Lobato, R, Boison, D, Clark, JD, et al. Opiate-induced changes in brain adenosine levels and narcotic drug responses. Neuroscience. (2013) 228:235–42. doi: 10.1016/j.neuroscience.2012.10.031

PubMed Abstract | Crossref Full Text | Google Scholar

137. Berrendero, F, Castane, A, Ledent, C, Parmentier, M, Maldonado, R, and Valverde, O. Increase of morphine withdrawal in mice lacking A2a receptors and no changes in CB1/A2a double knockout mice. Eur J Neurosci. (2003) 17:315–24. doi: 10.1046/j.1460-9568.2003.02439.x

PubMed Abstract | Crossref Full Text | Google Scholar

138. Beltran, NM, Parra, AN, Serrano, AP, Castillo, J, Castro, IM, Elsey, MK, et al. The effects of eating a traditional high fat/high carbohydrate or a ketogenic diet on sensitivity of female rats to morphine. J Pharmacol Exp Ther. (2024) 391:30–8. doi: 10.1124/jpet.124.002188

PubMed Abstract | Crossref Full Text | Google Scholar

139. Traina, G. The neurobiology of acetyl-L-carnitine. Front Biosci. (2016) 21:1314–29. doi: 10.2741/4459

PubMed Abstract | Crossref Full Text | Google Scholar

140. Crawford, J, Liu, S, Tao, R, Kramer, P, Bender, S, and Tao, F. The ketogenic diet mitigates opioid-induced hyperalgesia by restoring Short-chain fatty acids-producing Bacteria in the gut. Pain. (2024) 165:e106–14. doi: 10.1097/j.pain.0000000000003212

PubMed Abstract | Crossref Full Text | Google Scholar

141. Sethi, S, Sinha, A, and Gearhardt, AN. Low carbohydrate ketogenic therapy as a metabolic treatment for binge eating and Ultraprocessed food addiction. Curr Opin Endocrinol Diabetes Obes. (2020) 27:275–82. doi: 10.1097/MED.0000000000000571

PubMed Abstract | Crossref Full Text | Google Scholar

142. Carmen, M, Safer, DL, Saslow, LR, Kalayjian, T, Mason, AE, Westman, EC, et al. Treating binge eating and food addiction symptoms with low-carbohydrate ketogenic diets: a case series. J Eat Disord. (2020) 8:2. doi: 10.1186/s40337-020-0278-7

PubMed Abstract | Crossref Full Text | Google Scholar

143. Rostanzo, E, Marchetti, M, Casini, I, and Aloisi, AM. Very-low-calorie ketogenic diet: a potential treatment for binge eating and food addiction symptoms in women. A pilot study. Int J Environ Res Public Health. (2021) 18:12802. doi: 10.3390/ijerph182312802

PubMed Abstract | Crossref Full Text | Google Scholar

144. Danan, A, Westman, EC, Saslow, LR, and Ede, G. The ketogenic diet for refractory mental illness: a retrospective analysis of 31 inpatients. Front Psych. (2022) 13:951376. doi: 10.3389/fpsyt.2022.951376

PubMed Abstract | Crossref Full Text | Google Scholar

145. Laurent, N. Retrospective case study: ketogenic metabolic therapy in the effective Management of Treatment-Resistant Depressive Symptoms in bipolar disorder. Front Nutr. (2024) 11:1394679. doi: 10.3389/fnut.2024.1394679

PubMed Abstract | Crossref Full Text | Google Scholar

146. Freyberg, Z, Andreazza, A, McClung, C, and Phillips, ML. Linking mitochondrial dysfunction, neurotransmitter, neural network abnormalities and mania: elucidating neurobiological mechanisms of the therapeutic effect of the ketogenic diet in bipolar disorder. Biol Psychiatry Cogn Neurosci Neuroimag. (2024). doi: 10.1016/j.bpsc.2024.07.011

PubMed Abstract | Crossref Full Text | Google Scholar

147. Chrysafi, M, Jacovides, C, Papadopoulou, SK, Psara, E, Vorvolakos, T, Antonopoulou, M, et al. The potential effects of the ketogenic diet in the prevention and co-treatment of stress, anxiety, depression, schizophrenia, and bipolar disorder: from the basic research to the clinical practice. Nutrients. (2024) 16:1546. doi: 10.3390/nu16111546

PubMed Abstract | Crossref Full Text | Google Scholar

148. Calabrese, L, Frase, R, and Ghaloo, M. Complete remission of depression and anxiety using a ketogenic diet: case series. Front Nutr. (2024) 11:1396685. doi: 10.3389/fnut.2024.1396685

PubMed Abstract | Crossref Full Text | Google Scholar

149. Bellamy, EL, Hadjiefthyvoulou, F, Walsh, J, Brown, J, and Turner, J. Understanding the experiences of ketogenic metabolic therapy for people living with varying levels of depressive symptoms: a thematic analysis. Front Nutr. (2024) 11:1397546. doi: 10.3389/fnut.2024.1397546

Crossref Full Text | Google Scholar

150. Garner, S, Davies, E, Barkus, E, and Kraeuter, AK. Ketogenic diet has a positive association with mental and emotional well-being in the general population. Nutrition. (2024) 124:112420. doi: 10.1016/j.nut.2024.112420

PubMed Abstract | Crossref Full Text | Google Scholar

151. Sethi, S, Wakeham, D, Ketter, T, Hooshmand, F, Bjornstad, J, Richards, B, et al. Ketogenic diet intervention on metabolic and psychiatric health in bipolar and schizophrenia: a pilot trial. Psychiatry Res. (2024) 335:115866. doi: 10.1016/j.psychres.2024.115866

PubMed Abstract | Crossref Full Text | Google Scholar

152. Schweickart, A, Batra, R, Neth, BJ, Martino, C, Shenhav, L, Zhang, AR, et al. Serum and CSF metabolomics analysis shows Mediterranean ketogenic diet mitigates risk factors of Alzheimer's disease. NPJ Metab Health Dis. (2024) 2:15. doi: 10.1038/s44324-024-00016-3

PubMed Abstract | Crossref Full Text | Google Scholar

153. Ferre, S, O'Connor, WT, Snaprud, P, Ungerstedt, U, and Fuxe, K. Antagonistic interaction between adenosine A2A receptors and dopamine D2 receptors in the ventral Striopallidal system. Implications for the treatment of schizophrenia. Neuroscience. (1994) 63:765–73. doi: 10.1016/0306-4522(94)90521-5

PubMed Abstract | Crossref Full Text | Google Scholar

154. Boison, D, Singer, P, Shen, H-Y, Feldon, J, and Yee, BK. Adenosine hypothesis of schizophrenia – opportunities for pharmacotherapy. Neuropharmacology. (2012) 62:1527–43. doi: 10.1016/j.neuropharm.2011.01.048

PubMed Abstract | Crossref Full Text | Google Scholar

155. Shen, H-Y, Singer, P, Lytle, N, Wei, C, Lan, J-Q, Williams-Karnesky, RL, et al. Adenosine augmentation ameliorates psychotic and cognitive Endophenotypes of schizophrenia in mice. J Clin Invest. (2012) 122:2567–77. doi: 10.1172/JCI62378

PubMed Abstract | Crossref Full Text | Google Scholar

156. Lintunen, J, Lahteenvuo, M, Tiihonen, J, Tanskanen, A, and Taipale, H. Adenosine modulators and Calcium Channel blockers as add-on treatment for schizophrenia. NPJ Schizophr. (2021) 7:1. doi: 10.1038/s41537-020-00135-y

PubMed Abstract | Crossref Full Text | Google Scholar

157. Sahay, S, Devine, EA, McCullumsmith, RE, and O'Donovan, SM. Adenosine receptor mRNA expression in frontal cortical neurons in schizophrenia. Cells. (2023) 13:32. doi: 10.3390/cells13010032

PubMed Abstract | Crossref Full Text | Google Scholar

158. Marques, TR, Natesan, S, Rabiner, EA, Searle, GE, Gunn, R, Howes, OD, et al. Adenosine A2A receptor in schizophrenia: an in vivo brain PET imaging study. Psychopharmacology. (2022) 239:3439–45. doi: 10.1007/s00213-021-05900-0

PubMed Abstract | Crossref Full Text | Google Scholar

159. Townsend, L, Pillinger, T, Selvaggi, P, Veronese, M, Turkheimer, F, and Howes, O. Brain glucose metabolism in schizophrenia: a systematic review and Meta-analysis of 18FDG-PET studies in schizophrenia. Psychol Med. (2022) 53:4880–97. doi: 10.1017/S003329172200174X

PubMed Abstract | Crossref Full Text | Google Scholar

160. Kraeuter, AK, Archambault, N, van den Buuse, M, and Sarnyai, Z. Ketogenic diet and olanzapine treatment alone and in combination reduce a pharmacologically-induced Prepulse inhibition deficit in female mice. Schizophr Res. (2019) 212:221–4. doi: 10.1016/j.schres.2019.08.002

PubMed Abstract | Crossref Full Text | Google Scholar

161. Kraeuter, AK, Loxton, H, Lima, BC, Rudd, D, and Sarnyai, Z. Ketogenic diet reverses behavioral abnormalities in an acute NMDA receptor hypofunction model of schizophrenia. Schizophr Res. (2015) 169:491–3. doi: 10.1016/j.schres.2015.10.041

PubMed Abstract | Crossref Full Text | Google Scholar

162. Kraeuter, AK, Mashavave, T, Suvarna, A, van den Buuse, M, and Sarnyai, Z. Effects of Beta-Hydroxybutyrate administration on MK-801-induced schizophrenia-like behaviour in mice. Psychopharmacology. (2020) 237:1397–405. doi: 10.1007/s00213-020-05467-2

PubMed Abstract | Crossref Full Text | Google Scholar

163. Pacheco, A, Easterling, WS, and Pryer, MW. A pilot study of the ketogenic diet in schizophrenia. Am J Psychiatry. (1965) 121:1110–1. doi: 10.1176/ajp.121.11.1110

PubMed Abstract | Crossref Full Text | Google Scholar

164. Kraft, BD, and Westman, EC. Schizophrenia, gluten, and low-carbohydrate, ketogenic diets: a case report and review of the literature. Nutr Metab. (2009) 6:10. doi: 10.1186/1743-7075-6-10

PubMed Abstract | Crossref Full Text | Google Scholar

165. Palmer, CM, Gilbert-Jaramillo, J, and Westman, EC. The ketogenic diet and remission of psychotic symptoms in schizophrenia: two case studies. Schizophr Res. (2019) 208:439–40. doi: 10.1016/j.schres.2019.03.019

PubMed Abstract | Crossref Full Text | Google Scholar

166. Garner, S, Barkus, E, and Kraeuter, AK. Positive and negative Schizotypy personality traits are lower in individuals on ketogenic diet in a non-clinical Sample. Schizophr Res. (2024) 270:423–32. doi: 10.1016/j.schres.2024.07.010

PubMed Abstract | Crossref Full Text | Google Scholar

167. Anderson, RJ, Freedland, KE, Clouse, RE, and Lustman, PJ. The prevalence of comorbid depression in adults with diabetes: a Meta-analysis. Diabetes Care. (2001) 24:1069–78. doi: 10.2337/diacare.24.6.1069

PubMed Abstract | Crossref Full Text | Google Scholar

168. Lindekilde, N, Scheuer, SH, Rutters, F, Knudsen, L, Lasgaard, M, Rubin, KH, et al. Prevalence of type 2 diabetes in psychiatric disorders: an umbrella review with Meta-analysis of 245 observational studies from 32 systematic reviews. Diabetologia. (2022) 65:440–56. doi: 10.1007/s00125-021-05609-x

PubMed Abstract | Crossref Full Text | Google Scholar

169. Lustman, PJ, Anderson, RJ, Freedland, KE, de Groot, M, Carney, RM, and Clouse, RE. Depression and poor glycemic control: a Meta-analytic review of the literature. Diabetes Care. (2000) 23:934–42. doi: 10.2337/diacare.23.7.934

PubMed Abstract | Crossref Full Text | Google Scholar

170. Roy, T, and Lloyd, CE. Epidemiology of depression and diabetes: a systematic review. J Affect Disord. (2012) 142:S8–S21. doi: 10.1016/S0165-0327(12)70004-6

PubMed Abstract | Crossref Full Text | Google Scholar

171. Gonder-Frederick, LA, Cox, DJ, Bobbitt, SA, and Pennebaker, JW. Mood changes associated with blood glucose fluctuations in insulin-dependent diabetes mellitus. Health Psychol. (1989) 8:45–59. doi: 10.1037/0278-6133.8.1.45

PubMed Abstract | Crossref Full Text | Google Scholar

172. Laaboub, N, Locatelli, I, Grosu, C, Piras, M, Ngoc, TH, Ranjbar, S, et al. Metabolic disturbances are risk factors for readmission to psychiatric hospitals in non-smokers but not in smokers: results from a Swiss psychiatric cohort and in first-episode psychosis patients. Front Psych. (2024) 15:1256416. doi: 10.3389/fpsyt.2024.1256416

PubMed Abstract | Crossref Full Text | Google Scholar

173. Perry, BI, Stochl, J, Upthegrove, R, Zammit, S, Wareham, N, Langenberg, C, et al. Longitudinal trends in childhood insulin levels and body mass index and associations with risks of psychosis and depression in young adults. JAMA Psychiatry. (2021) 78:416–25. doi: 10.1001/jamapsychiatry.2020.4180

PubMed Abstract | Crossref Full Text | Google Scholar

174. Cox, DJ, McCall, A, Kovatchev, B, Sarwat, S, Ilag, LL, and Tan, MH. Effects of blood glucose rate of changes on perceived mood and cognitive symptoms in insulin-treated type 2 diabetes. Diabetes Care. (2007) 30:2001–2. doi: 10.2337/dc06-2480

PubMed Abstract | Crossref Full Text | Google Scholar

175. Kovatchev, B, Cox, DJ, Summers, KH, Gonder-Frederick, LA, and Clarke, WL. Postprandial glucose dynamics and associated symptoms in type 2 diabetes mellitus. J Appl Res. (2003) 4:449–58.

Google Scholar

176. Penckofer, S, Quinn, L, Byrn, M, Ferrans, C, Miller, M, and Strange, P. Does glycemic variability impact mood and quality of life? Diabetes Technol Ther. (2012) 14:303–10. doi: 10.1089/dia.2011.0191

PubMed Abstract | Crossref Full Text | Google Scholar

177. Muijs, LT, Racca, C, de Wit, M, Brouwer, A, Wieringa, TH, de Vries, R, et al. Glucose variability and mood in adults with diabetes: a systematic review. Endocrinol Diabetes Metab. (2021) 4:e00152. doi: 10.1002/edm2.152

PubMed Abstract | Crossref Full Text | Google Scholar

178. Reddy, M, Godsland, IF, Barnard, KD, Herrero, P, Georgiou, P, Thomson, H, et al. Glycemic variability and its impact on quality of life in adults with type 1 diabetes. J Diabetes Sci Technol. (2015) 10:60–6. doi: 10.1177/1932296815601440

PubMed Abstract | Crossref Full Text | Google Scholar

179. Knuppel, A, Shipley, MJ, Llewellyn, CH, and Brunner, EJ. Sugar intake from sweet food and beverages, common mental disorder and depression: prospective findings from the Whitehall II study. Sci Rep. (2017) 7:6287. doi: 10.1038/s41598-017-05649-7

PubMed Abstract | Crossref Full Text | Google Scholar

180. Castro, A, Gili, M, Visser, M, Penninx, B, Brouwer, IA, Montano, JJ, et al. Soft drinks and symptoms of depression and anxiety in overweight subjects: a longitudinal analysis of an European cohort. Nutrients. (2023) 15:3865. doi: 10.3390/nu15183865

PubMed Abstract | Crossref Full Text | Google Scholar

181. Chen, H, Cao, Z, Hou, Y, Yang, H, Wang, X, and Xu, C. The associations of dietary patterns with depressive and anxiety symptoms: a prospective study. BMC Med. (2023) 21:307. doi: 10.1186/s12916-023-03019-x

PubMed Abstract | Crossref Full Text | Google Scholar

182. Campayo, A, de, P, Roy, JF, Saz, P, de, C, Quintanilla, MA, et al. Depressive disorder and incident diabetes mellitus: the effect of characteristics of depression. Am J Psychiatry. (2010) 167:580–8. doi: 10.1176/appi.ajp.2009.09010038

PubMed Abstract | Crossref Full Text | Google Scholar

183. Carnethon, MR, Biggs, ML, Barzilay, JI, Smith, NL, Vaccarino, V, Bertoni, AG, et al. Longitudinal association between depressive symptoms and incident type 2 diabetes mellitus in older adults: the cardiovascular health study. Arch Intern Med. (2007) 167:802–7. doi: 10.1001/archinte.167.8.802

Crossref Full Text | Google Scholar

184. Bai, L, Zhou, Y, Zhang, J, and Ma, J. The role of a ketogenic diet in the treatment of dementia in type 2 diabetes mellitus. Nutrients. (2023) 15:1971. doi: 10.3390/nu15081971

PubMed Abstract | Crossref Full Text | Google Scholar

185. Di Lucente, J, Persico, G, Zhou, Z, Jin, L-W, Ramsey, J, Rutkowsky, J, et al. Ketogenic diet and BHB rescue the fall of long-term potentiation in an Alzheimer’s mouse model and stimulates synaptic plasticity pathway enzymes. Commun Biol. (2024) 7:195. doi: 10.1038/s42003-024-05860-z

PubMed Abstract | Crossref Full Text | Google Scholar

186. Rong, L, Peng, Y, Shen, Q, Chen, K, Fang, B, and Li, W. Effects of ketogenic diet on cognitive function of patients with Alzheimer's disease: a systematic review and Meta-analysis. J Nutr Health Aging. (2024) 28:100306. doi: 10.1016/j.jnha.2024.100306

PubMed Abstract | Crossref Full Text | Google Scholar

187. Buchholtz, A, Deme, P, Betz, JF, Brandt, J, Haughey, N, and Cervenka, MC. A randomized feasibility trial of the modified Atkin diet in older adults with mild cognitive impairment due to Alzheimer's disease. Front Endocrinol. (2024) 15:1182519. doi: 10.3389/fendo.2024.1182519

PubMed Abstract | Crossref Full Text | Google Scholar

188. Bosworth, A, Loh, V, Stranahan, BN, and Palmer, CM. Case report: ketogenic diet acutely improves cognitive function in patient with down syndrome and Alzheimer's disease. Front Psych. (2023) 13:1085512. doi: 10.3389/fpsyt.2022.1085512

PubMed Abstract | Crossref Full Text | Google Scholar

189. Phillips, MCL, Deprez, LM, Mortimer, GMN, Murtagh, DKJ, McCoy, S, Mylchreest, R, et al. Randomized crossover trial of a modified ketogenic diet in Alzheimer's disease. Alzheimers Res Ther. (2021) 13:51. doi: 10.1186/s13195-021-00783-x

PubMed Abstract | Crossref Full Text | Google Scholar

190. Krikorian, R, Shidler, MD, Summer, SS, Sullivan, PG, Duker, AP, Isaacson, RS, et al. Nutritional ketosis for mild cognitive impairment in Parkinson's disease: a controlled pilot trial. Clin Park Relat Disord. (2019) 1:41–7. doi: 10.1016/j.prdoa.2019.07.006

PubMed Abstract | Crossref Full Text | Google Scholar

191. Taylor, MK, Sullivan, DK, Mahnken, JD, Burns, JM, and Swerdlow, RH. Feasibility and efficacy data from a ketogenic diet intervention in Alzheimer's disease. Alzheimers Dement (NY). (2018) 4:28–36. doi: 10.1016/j.trci.2017.11.002

PubMed Abstract | Crossref Full Text | Google Scholar

192. Ota, M, Matsuo, J, Ishida, I, Takano, H, Yokoi, Y, Hori, H, et al. Effects of a medium-chain triglyceride-based ketogenic formula on cognitive function in patients with mild-to-moderate Alzheimer's disease. Neurosci Lett. (2019) 690:232–6. doi: 10.1016/j.neulet.2018.10.048

PubMed Abstract | Crossref Full Text | Google Scholar

193. Reger, MA, Henderson, ST, Hale, K, Cholerton, B, Baker, LD, Watson, GS, et al. Effects of β-Hydroxybutyrate on cognition in memory-impaired adults. Neurobiol Aging. (2004) 25:311–4. doi: 10.1016/S0197-4580(03)00087-3

PubMed Abstract | Crossref Full Text | Google Scholar

194. Henderson, ST, Vogel, JL, Barr, LJ, Garvin, F, Jones, JJ, and Constantini, LC. Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer's disease: a randomized, double-blind, placebo-controlled, multicenter trial. Nutr Metab. (2009) 6:31. doi: 10.1186/1743-7075-6-31

PubMed Abstract | Crossref Full Text | Google Scholar

195. Fortier, M, Castellano, CA, St-Pierre, V, Myette-Côté, É, Langlois, F, Roy, M, et al. A ketogenic drink improves cognition in mild cognitive impairment: results of a 6-month RCT. Alzheimers Dement. (2021) 17:543–52. doi: 10.1002/alz.12206

PubMed Abstract | Crossref Full Text | Google Scholar

196. Roy, M, Fortier, M, Rheault, F, Edde, M, Croteau, E, Castellano, CA, et al. A ketogenic supplement improves white matter energy supply and processing speed in mild cognitive impairment. Alzheimers Dement (NY). (2021) 7:e12217. doi: 10.1002/trc2.12217

PubMed Abstract | Crossref Full Text | Google Scholar

197. Fortier, M, Castellano, CA, Croteau, E, Langlois, F, Bocti, C, St-Pierre, V, et al. A ketogenic drink improves brain energy and some measures of cognition in mild cognitive impairment. Alzheimers Dement. (2019) 15:625–34. doi: 10.1016/j.jalz.2018.12.017

PubMed Abstract | Crossref Full Text | Google Scholar

198. Zilbovicius, M, Boddaert, N, Belin, P, Poline, JB, Remy, P, Mangin, JF, et al. Temporal lobe dysfunction in childhood autism: a PET study. Positron emission tomography. Am J Psychiatry. (2000) 157:1988–93. doi: 10.1176/appi.ajp.157.12.1988

PubMed Abstract | Crossref Full Text | Google Scholar

199. Ohnishi, T, Matsuda, H, Hashimoto, T, Kunihiro, T, Nishikawa, M, Uema, T, et al. Abnormal regional cerebral blood flow in childhood autism. Brain. (2000) 123:1838–44. doi: 10.1093/brain/123.9.1838

PubMed Abstract | Crossref Full Text | Google Scholar

200. Jann, K, Hernandez, LM, Beck-Pancer, D, McCarron, R, Smith, RX, Dapretto, M, et al. Altered resting perfusion and functional connectivity of default mode network in youth with autism Spectrum disorder. Brain Behav. (2015) 5:e00358. doi: 10.1002/brb3.358

PubMed Abstract | Crossref Full Text | Google Scholar

201. Jendle, J, Agvall, B, Galozy, A, and Adolfsson, P. Patterns and predictors associated with long-term glycemic control in pediatric and young adult patients with type 1 diabetes. J Diabetes Sci Technol. (2023) 17:1243–51. doi: 10.1177/19322968221096423

PubMed Abstract | Crossref Full Text | Google Scholar

202. Cheng, N, Rho, JM, and Masino, SA. Metabolic dysfunction underlying autism spectrum disorder and potential treatment approaches. Front Mol Neurosci. (2017) 10:34. doi: 10.3389/fnmol.2017.00034

Crossref Full Text | Google Scholar

203. Masino, SA, Kawamura, M Jr, Cote, JL, Williams, RB, and Ruskin, DN. Adenosine and autism: a Spectrum of opportunities. Neuropharmacology. (2013) 68:116–21. doi: 10.1016/j.neuropharm.2012.08.013

PubMed Abstract | Crossref Full Text | Google Scholar

204. Masino, SA, Kawamura, M Jr, Plotkin, LM, Svedova, J, DiMario, FJ, and Eigsti, IM. The relationship between the neuromodulator adenosine and behavioral symptoms of autism. Neurosci Lett. (2011) 500:1–5. doi: 10.1016/j.neulet.2011.06.007

PubMed Abstract | Crossref Full Text | Google Scholar

205. Verpeut, JL, DiCicco-Bloom, E, and Bello, NT. Ketogenic diet exposure during the juvenile period increases social behaviors and forebrain neural activation in adult engrailed 2 null mice. Physiol Behav. (2016) 161:90–8. doi: 10.1016/j.physbeh.2016.04.001

PubMed Abstract | Crossref Full Text | Google Scholar

206. Dai, Y, Zhao, Y, Tomi, S, Shin, B-C, Thamotharan, S, Mazarati, A, et al. Sex-specific life course changes in the neuro-metabolic phenotype of Glut3 null heterozygous mice: ketogenic diet ameliorates electroencephalographic seizures and improves sociability. Endocrinology. (2017) 158:936–49. doi: 10.1210/en.2016-1816

PubMed Abstract | Crossref Full Text | Google Scholar

207. Mu, C, Corley, MJ, Lee, RWY, Wong, M, Pang, A, Arakaki, G, et al. Metabolic framework for the improvement of autism spectrum disorders by a modified ketogenic diet: a pilot study. J Proteome Res. (2020) 19:382–90. doi: 10.1021/acs.jproteome.9b00581

PubMed Abstract | Crossref Full Text | Google Scholar

208. Spilioti, M, Evangeliou, AE, Tramma, D, Theodoridou, Z, Metaxas, S, Michailidi, E, et al. Evidence for treatable inborn errors of metabolism in a cohort of 187 Greek patients with autism Spectrum disorder (ASD). Front Hum Neurosci. (2013) 7:858. doi: 10.3389/fnhum.2013.00858

PubMed Abstract | Crossref Full Text | Google Scholar

209. Żarnowska, I, Chrapko, B, Gwizda, G, Nocuń, A, Mitosek-Szewczyk, K, and Gasior, M. Therapeutic use of carbohydrate-restricted diets in an autistic child; a case report of clinical and 18FDG PET findings. Metab Brain Dis. (2018) 33:1187–92. doi: 10.1007/s11011-018-0219-1

PubMed Abstract | Crossref Full Text | Google Scholar

210. Lee, RWY, Corley, MJ, Pang, A, Arakaki, G, Abbott, L, Nishimoto, M, et al. A modified ketogenic gluten-free diet with MCT improves behavior in children with autism Spectrum disorder. Physiol Behav. (2018) 188:205–11. doi: 10.1016/j.physbeh.2018.02.006

PubMed Abstract | Crossref Full Text | Google Scholar

211. Herbert, MR, and Buckley, JA. Autism and dietary therapy: case report and review of the literature. J Child Neurol. (2013) 28:975–82. doi: 10.1177/0883073813488668

PubMed Abstract | Crossref Full Text | Google Scholar

212. Evangeliou, A, Vlachonikolis, I, Mihailidou, H, Spilioti, M, Skarpalezou, A, Makaronas, N, et al. Application of a ketogenic diet in children with autistic behavior: pilot study. J Child Neurol. (2003) 18:113–8. doi: 10.1177/08830738030180020501

PubMed Abstract | Crossref Full Text | Google Scholar

213. El-Rashidy, O, El-Baz, F, El-Gendy, Y, Khalaf, R, Reda, D, and Saad, K. Ketogenic diet versus gluten free casein free diet in autistic children: a case-control study. Metab Brain Dis. (2017) 32:1935–41. doi: 10.1007/s11011-017-0088-z

PubMed Abstract | Crossref Full Text | Google Scholar

214. Alluwyam, AH, and Estrella, ED. Ketogenic diet and its potential role in preventing type 2 diabetes mellitus and its complications: a narrative review of randomized controlled trials. Cureus. (2024) 16:e66419. doi: 10.7759/cureus.66419

PubMed Abstract | Crossref Full Text | Google Scholar

215. Katyal, NG, Koehler, AN, McGhee, B, Foley, CM, and Crumrine, PK. The ketogenic diet in refractory epilepsy: the experience of Children's Hospital of Pittsburgh. Clin Pediatr (Phila). (2000) 39:153–9. doi: 10.1177/000992280003900303

PubMed Abstract | Crossref Full Text | Google Scholar

216. Hemingway, C, Freeman, JM, Pillas, DJ, and Pyzik, PL. The ketogenic diet: a 3- to 6-year follow-up of 150 children enrolled prospectively. Pediatrics. (2001) 108:898–905. doi: 10.1542/peds.108.4.898

PubMed Abstract | Crossref Full Text | Google Scholar

217. MacCracken, KA, and Scalisi, JC. Development and evaluation of a ketogenic diet program. J Am Diet Assoc. (1999) 99:1554–8. doi: 10.1016/S0002-8223(99)00381-8

PubMed Abstract | Crossref Full Text | Google Scholar

218. Nabbout, R, Copioli, C, Chipaux, M, Chemaly, N, Desguerre, I, Dulac, O, et al. Ketogenic diet also benefits Dravet syndrome patients receiving Stiripentol: a prospective pilot study. Epilepsia. (2011) 52:e54–7. doi: 10.1111/j.1528-1167.2011.03107.x

PubMed Abstract | Crossref Full Text | Google Scholar

219. Yilmaz, U, Edizer, S, Akisin, Z, Kose, M, Guzin, Y, Gurbuz, G, et al. The effectiveness of the ketogenic diet in drug-resistant childhood epilepsy. Turk J Pediatr. (2022) 64:210–20. doi: 10.24953/turkjped.2021.4

PubMed Abstract | Crossref Full Text | Google Scholar

220. Budoff, M, Manubolu, VS, Kinninger, A, Norwitz, NG, Feldman, D, Wood, TR, et al. Carbohydrate restriction-induced elevations in LDL-cholesterol and atherosclerosis: the keto trial. JACC Adv. (2024) 3:101109. doi: 10.1016/j.jacadv.2024.101109

PubMed Abstract | Crossref Full Text | Google Scholar

221. Diamond, DM, Mason, P, and Bikman, BT. Opinion: are mental health benefits of the ketogenic diet accompanied by an increased risk of cardiovascular disease? Front Nutr. (2024) 11:1394610. doi: 10.3389/fnut.2024.1394610

PubMed Abstract | Crossref Full Text | Google Scholar

222. Diamond, D, Bikman, B, and Mason, P. Statin therapy is not warranted for a person with high LDL-cholesterol on a low-carbohydrate diet. Curr Opin Endocrinol Diabetes Obes. (2022) 29:497–511. doi: 10.1097/MED.0000000000000764

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: ketogenic diet, adenosine, dopamine, psychiatric disorder, addiction, mental health, metabolic psychiatry

Citation: Ruskin DN, Martinez LA and Masino SA (2025) Ketogenic diet, adenosine, and dopamine in addiction and psychiatry. Front. Nutr. 12:1492306. doi: 10.3389/fnut.2025.1492306

Received: 06 September 2024; Accepted: 11 February 2025;
Published: 10 March 2025.

Edited by:

Ramón Sotomayor-Zárate, Universidad de Valparaiso, Chile

Reviewed by:

José Luis Marcos, Viña del Mar University, Chile

Copyright © 2025 Ruskin, Martinez and Masino. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: David N. Ruskin, ZGF2aWQucnVza2luQHRyaW5jb2xsLmVkdQ==; Susan A. Masino, c3VzYW4ubWFzaW5vQHRyaW5jb2xsLmVkdQ==

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

Research integrity at Frontiers

94% of researchers rate our articles as excellent or good

Learn more about the work of our research integrity team to safeguard the quality of each article we publish.


Find out more