1
|
Ruskin DN, Martinez LA, Masino SA. Ketogenic diet, adenosine, and dopamine in addiction and psychiatry. Front Nutr 2025; 12:1492306. [PMID: 40129664 PMCID: PMC11932665 DOI: 10.3389/fnut.2025.1492306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/11/2025] [Indexed: 03/26/2025] Open
Abstract
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.
Collapse
|
2
|
Li X, Young AJ, Shi Z, Byanyima J, Vesslee S, Reddy R, Pond T, Elliott M, Reddy R, Doot RK, van der Veen JW, Kranzler HR, Reddy Nanga RP, Dubroff JG, Wiers CE. Pharmacokinetic effects of a single-dose nutritional ketone ester supplement on brain glucose and ketone metabolism in alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2023.09.25.23296090. [PMID: 37808798 PMCID: PMC10557835 DOI: 10.1101/2023.09.25.23296090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Acute alcohol intake decreases brain glucose metabolism and increases brain uptake of acetate, a metabolite of alcohol. This shift in energy utilization persists beyond acute intoxication in individuals with alcohol use disorder (AUD), and may contribute to alcohol craving. We recently found that ketone therapies decrease alcohol withdrawal and alcohol craving in AUD. Here, we studied the effects of a single-dose ketone ester (KE) supplement on brain energy metabolism and alcohol craving. Five AUD and five healthy control (HC) participants underwent two 18 F-fluorodeoxyglucose positron emission tomography (PET) scans, after consumption of 395 mg/kg KE or without (baseline), in randomized order. In the AUD group, KE reduced alcohol craving scores compared to baseline. KE decreased blood glucose levels and elevated blood β-hydroxybutyrate (BHB) levels compared to baseline in both groups. Whole-brain voxel-wise maps of the cerebral metabolic rate of glucose (CMRglc) decreased by 17% in both groups, with the largest KE-induced CMRglc reductions in the frontal, occipital, and cingulate cortices, hippocampus, amygdala, and insula. There were no group differences between AUD and HC in blood or FDG measures, and no correlations between reductions in craving with CMRglc. Cingulate BHB levels, as assessed with 1 H-magnetic resonance spectroscopy in 5 participant with AUD, increased 3-fold with KE compared to baseleline. In sum, administration of a single dose of KE rapidly shifted brain energetics from glucose to ketone metabolism in HC and AUD. KE also reduced ratings of alcohol craving, demonstrating its potential clinical effectiveness for supporting brain health and alcohol craving in AUD.
Collapse
|
3
|
Pereira-Rufino LDS, Gobbo DR, Conte R, Sino RMD, Oliveira NND, Fidalgo TM, Sato JR, Carrete Junior H, Souza-Formigoni MLO, Shi Z, Vissoci JRN, Wiers CE, Céspedes IC. Reduced gray matter volume in limbic and cortical areas is associated with anxiety and depression in alcohol use disorder patients. Psychiatry Res Neuroimaging 2025; 347:111946. [PMID: 39823711 DOI: 10.1016/j.pscychresns.2025.111946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/20/2025]
Abstract
Alcohol use disorder (AUD) is a multifactorial disease closely related to neurodevelopment and environmental factors that influence behavior. This study explored the relationships between brain volume and behavior from an Exploratory Structural Equation Modeling (ESEM) based on the Research Domain Criteria. High-resolution magnetic resonance imaging scans were acquired from recent patients with AUD (n = 50) and healthy controls (HC=50). Group differences were assessed by means of voxel-based morphometry (VBM) and regions of interest (ROIs). Participants completed a battery of neurocognitive tasks and emotional tests. When controlling for age, education levels, and total intracranial volume, we found lower gray matter volume in cortical and limbic areas, as well as significant impairments in the AUD group on cognition and affective status. This study demonstrated the importance of multifactorial analysis and complex models in order to better understand substance use disorders. The brain losses may have been the result of neurodevelopmental impairments due to biological and environmental factors that predisposed to AUD or the result of drug abuse. The ESEM indicated that limbic areas indirectly affected the alcohol severity through emotional dysfunction. These results suggest a more relevant involvement of limbic regions for the severity of alcohol use, showing a more significant association between AUD and mood disorders.
Collapse
Affiliation(s)
- Laís da Silva Pereira-Rufino
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Denise Ribeiro Gobbo
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Rafael Conte
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - Raissa Mazzer de Sino
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | | | - Thiago Marques Fidalgo
- Departamento de Psiquiatria, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil
| | - João Ricardo Sato
- Interdisciplinary Unit for Applied Neuroscience (NINA) / Center for Mathematics, Computing and Cognition (CMCC), Universidade Federal do ABC - UFABC, São Bernardo do Campo, SP, Brazil
| | - Henrique Carrete Junior
- Departamento de Diagnóstico por Imagem, Escola Paulista de Medicina, Universidade Federal de São Paulo, UNIFESP, São Paulo, SP, Brazil
| | | | - Zhenhao Shi
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Corinde E Wiers
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Isabel Cristina Céspedes
- Departamento de Morfologia e Genética, Escola Paulista de Medicina, Universidade Federal de São Paulo - UNIFESP, São Paulo, SP, Brazil.
| |
Collapse
|
4
|
Ehinger Y, Laguesse S, Phamluong K, Salvi A, Sei YJ, Hoisington ZW, Soneja D, Gunasekaran S, Nakamura K, Ron D. Paradoxical mTORC1-Dependent microRNA-mediated Translation Repression in the Nucleus Accumbens of Mice Consuming Alcohol Attenuates Glycolysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.29.569312. [PMID: 38076984 PMCID: PMC10705386 DOI: 10.1101/2023.11.29.569312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
mTORC1 promotes protein translation, learning and memory, and neuroadaptations that underlie alcohol use and abuse. We report that activation of mTORC1 in the nucleus accumbens (NAc) of mice consuming alcohol promotes the translation of microRNA (miR) machinery components and the upregulation of microRNAs (miRs) expression including miR-34a-5p. In parallel, we detected a paradoxical mTORC1-dependent repression of translation of transcripts including Aldolase A, an essential glycolytic enzyme. We found that miR-34a-5p in the NAc targets Aldolase A for translation repression and promotes alcohol intake. Our data further suggest that glycolysis is inhibited in the NAc manifesting in an mTORC1-dependent attenuation of L-lactate, the end product of glycolysis. Finally, we show that systemic administration of L-lactate attenuates mouse excessive alcohol intake. Our data suggest that alcohol promotes paradoxical actions of mTORC1 on translation and glycolysis which in turn drive excessive alcohol use.
Collapse
|
5
|
Ji J, Tang Y. A ketogenic diet regulates microglial activation to treat drug addiction. Front Pharmacol 2025; 16:1462699. [PMID: 39917617 PMCID: PMC11799558 DOI: 10.3389/fphar.2025.1462699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
Drug addiction is a chronic and potentially deadly disease that is considered a global health problem and describes the alteration of brain function by psychostimulant drugs through changes in the reward system. However, there is still no ideal strategy for the management of drug addiction. Previous studies have suggested that microglia are involved in events associated with neuroplasticity and memory, which are also related to drug addiction. Many studies have shown that psychoactive substances may act directly on immune cells, altering their function and inducing the production of various inflammatory mediators. In recent years, a ketogenic diet (KD) was shown to have therapeutic benefits as a dietary therapy for a variety of neurological disorders. With respect to drug addiction, studies have shown that a KD can alleviate glucose metabolism disorders caused by alcohol use disorders by increasing ketone metabolism, thereby reducing withdrawal symptoms. This finding indicates the potential of a KD as a treatment for drug addiction, since a KD may promote the transition of microglia to a predominantly anti-inflammatory state through several mechanisms. Here, we discuss recent research showing that a KD plays a variety of roles in controlling microglia-mediated inflammation, opening new treatment avenues to treat drug addiction. This succinct analysis offers evidence of the enormous potential of a KD to treat drug addiction through the inhibition of microglial activation.
Collapse
Affiliation(s)
- Jie Ji
- Department of Comprehensive (VIP) Inpatient Ward, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Yousufzai W, Singh M, Ahmadi L, Balamurali S, Bavishi D, Ashraf S, Stuart DB, Baronia R, Amor W. Measuring the effects of ketogenic diet on neuropsychiatric disorder: A scoping review. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111205. [PMID: 39592022 DOI: 10.1016/j.pnpbp.2024.111205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVE This scoping review aims to examine the available literature on the ketogenic diet's (KD) efficiency as a potential therapeutic intervention for various neuropsychiatric disorders. INTRODUCTION The KD is a high-fat, low-carbohydrate diet that has been studied for its potential benefits in managing neuropsychiatric disorders. However, the extent of its effectiveness across a spectrum of these conditions remains unclear. INCLUSION CRITERIA The study designs considered eligible encompassed randomized and non-randomized controlled trials, retrospective and prospective observational studies, and comparative effectiveness assessments. The criteria for including each study were specifically related to neuropsychiatric disorders, referring to the DSM-5 coding guidelines. METHODS A systemic search was performed by an experienced reference librarian across multiple databases to pinpoint studies relevant to the influence of the ketogenic diet on neuropsychiatric disorders. All relevant articles were included that ranged over the last thirteen years. All relevant records identified were compiled into the Covidence systematic review software. RESULTS A total of 30 studies were reviewed, which reported effects of the KD on neuropsychiatric disorders, including improvements in Global Developmental Delay, Childhood Autism, Attention Deficit/Hyperactivity Disorder (ADHD) symptoms, psychotic symptoms, Bipolar and Related Disorders, Depressive Disorder symptoms, anxiety symptoms, eating disorders, Substance-Related and Addictive Disorders, Major and Mild Neurocognitive Disorders, and Seizure Disorders. CONCLUSION The KD may serve as a promising therapeutic intervention for various neuropsychiatric disorders. However, the evidence is heterogeneous, and further rigorous research is needed to establish the KD as a standard treatment for these disorders and to understand the underlying mechanisms of its effects. IMPLICATIONS FOR PRACTICE This review underscores the need for healthcare professionals to consider the potential benefits and limitations of the KD when managing patients with neuropsychiatric disorders. It also highlights the importance of individualized treatment plans based on the specific needs and responses of each patient.
Collapse
Affiliation(s)
- Wali Yousufzai
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| | - Monika Singh
- Himalayan Institute of Medical Science, Uttarakhand, India
| | | | - Shreya Balamurali
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| | - Divyaraj Bavishi
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| | - Sahar Ashraf
- Texas Tech University Health Sciences Center, Permian Basin, Midland, TX, USA.
| | - Daniel B Stuart
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| | - Regina Baronia
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| | - Wail Amor
- Psychiatry Department, Texas Tech University Health Sciences Center, 3601 4th Street STOP 8103, Lubbock, TX 79430-8103, USA.
| |
Collapse
|
7
|
Wang X, Manza P, Li X, Ramos‐Rolón A, Hager N, Wang G, Volkow ND, Hu Y, Shi Z, Wiers CE. Reduced brain network segregation in alcohol use disorder: Associations with neurocognition. Addict Biol 2024; 29:e13446. [PMID: 39686721 PMCID: PMC11649955 DOI: 10.1111/adb.13446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 12/18/2024]
Abstract
The human brain consists of functionally segregated networks, characterized by strong connections among regions belonging to the same network and weak connections between those of different networks. Alcohol use disorder (AUD) is associated with premature brain aging and neurocognitive impairments. Given the link between decreased brain network segregation and age-related cognitive decline, we hypothesized lower brain segregation in patients with AUD than healthy controls (HCs). Thirty AUD patients (9 females, 21 males) and 61 HCs (35 females, 26 males) underwent resting-state functional MRI (rs-fMRI), whose data were processed to assess segregation within the brain sensorimotor and association networks. We found that, compared to HCs, AUD patients had significantly lower segregation in both brain networks as well as poorer performance on a spatial working memory task. In the HC group, brain network segregation correlated negatively with age and positively with spatial working memory. Our findings suggest reduced brain network segregation in individuals with AUD that may contribute to cognitive impairment and is consistent with premature brain aging in this population.
Collapse
Affiliation(s)
- Xinying Wang
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Psychology and Behavioral SciencesZhejiang University, Zijingang CampusHangzhouZhejiang ProvinceChina
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMarylandUSA
| | - Xinyi Li
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Astrid Ramos‐Rolón
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Nathan Hager
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Gene‐Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMarylandUSA
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthBethesdaMarylandUSA
| | - Yuzheng Hu
- Department of Psychology and Behavioral SciencesZhejiang University, Zijingang CampusHangzhouZhejiang ProvinceChina
| | - Zhenhao Shi
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Corinde E. Wiers
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
8
|
Edwards MGP, Furuholmen-Jenssen T, Søegaard EGI, Thapa SB, Andersen JR. Exploring diet-induced ketosis with exogenous ketone supplementation as a potential intervention in post-traumatic stress disorder: a feasibility study. Front Nutr 2024; 11:1406366. [PMID: 39588043 PMCID: PMC11586679 DOI: 10.3389/fnut.2024.1406366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 11/27/2024] Open
Abstract
Background Post-Traumatic Stress Disorder (PTSD) is a severe and pervasive mental disorder, and patients experience numerous distressing symptoms and impairments that significantly impact their lives. In addition to being a mental disorder, PTSD is strongly associated with a wide range of metabolic abnormalities that affect the entire body. Existing treatment options of psychotherapy and medications are often ineffective. Exploring other potential treatments is necessitated. The ketogenic diet has shown potential as a metabolic therapy in certain neurological and mental disorders and is a promising intervention in the treatment of PTSD. Aim This study aimed to examine if a 4-week ketogenic diet intervention supplemented with exogenous ketones (KD-KS) was feasible in adult patients with PTSD, to what extent it was possible to recruit patients, attain and maintain ketosis (plasma concentration of β-hydroxybutyrate (BHB) ≥ 0.5 mmol/L), the occurrence of serious adverse reactions and adverse reactions to KD-KS, and acceptance of treatment. Our exploratory aims were changes in PTSD symptoms and health-related quality of life (QoL) from baseline to 4 weeks. Methods Patients 18 ≤ 65 years old, diagnosed with PTSD, and receiving outpatient treatment for PTSD at Southern Oslo District Psychiatric Centre (DPC), Oslo University Hospital, Oslo, Norway, were included. The intervention consisted of a ketogenic diet supplemented with β-hydroxybutyrate salt to obtain ketosis. PTSD symptoms were measured with the PTSD Checklist for DSM-5 (PCL-5) and QoL was measured with the RAND 36-Item Health Survey 1.0. Results During a 21-week inclusion period, three of four eligible patients (75% [95% CI: 30 to 95%]) were included. Two patients (67% [95% CI: 21 to 94%]) completed the 4-week intervention and one patient (33% [95% CI: 6 to 79%]) completed 2 weeks of intervention before discontinuing. Ketosis was achieved on day 1 in one patient, and on day 2 in two patients, and was maintained in 87% of the intervention. There were no serious adverse reactions. Adverse reactions were reported in a total of 70% of intervention days, the most frequent being headache followed by fatigue. The participant-perceived degree of adverse reactions was low to moderate. The treatment was accepted by patients on all intervention days. PCL-5 decreased by 20 points (70 to 50) in patient 1 and by 10 points (50 to 40) in patient 2, from baseline to 4 weeks, which is a reliable and clinically meaningful improvement. QoL improved in six of eight RAND-36 subscales in patient 1 and three of eight in patient 2. Patient 3 did not complete assessments after week 2. Conclusion To the best of our knowledge, this feasibility study is the first study examining a ketogenic diet intervention in patients with PTSD. Three of four predefined feasibility criteria were achieved. Ketosis was attained fast and maintained, patients were compliant and there were clinically meaningful improvements in PTSD symptoms and QoL. Despite the small sample size, the knowledge obtained in this study is important for the planning of future studies with ketogenic diet interventions in this patient group. It is a first step for potential dietary and metabolic therapies in PTSD. Further feasibility and pilot studies with larger sample sizes are needed to determine feasibility and safety before planning future randomised controlled trials investigating an effect. Clinical trial registration https://ClinicalTrials.gov, identifier NCT05415982.
Collapse
Affiliation(s)
- Maria G. P. Edwards
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Tobias Furuholmen-Jenssen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Erik Ganesh Iyer Søegaard
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Suraj Bahadur Thapa
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
- Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Jens R. Andersen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Torres-Rubio L, Reguilón MD, Mellado S, Pascual M, Rodríguez-Arias M. Effects of Ketogenic Diet on Increased Ethanol Consumption Induced by Social Stress in Female Mice. Nutrients 2024; 16:2814. [PMID: 39275131 PMCID: PMC11397041 DOI: 10.3390/nu16172814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Stress is a critical factor in the development of mental disorders such as addiction, underscoring the importance of stress resilience strategies. While the ketogenic diet (KD) has shown efficacy in reducing alcohol consumption in male mice without cognitive impairment, its impact on the stress response and addiction development, especially in females, remains unclear. This study examined the KD's effect on increasing ethanol intake due to vicarious social defeat (VSD) in female mice. Sixty-four female OF1 mice were divided into two dietary groups: standard diet (n = 32) and KD (n = 32). These were further split based on exposure to four VSD or exploration sessions, creating four groups: EXP-STD (n = 16), VSD-STD (n = 16), EXP-KD (n = 16), and VSD-KD (n = 16). KD-fed mice maintained ketosis from adolescence until the fourth VSD/EXP session, after which they switched to a standard diet. The Social Interaction Test was performed 24 h after the last VSD session. Three weeks post-VSD, the Drinking in the Dark test and Oral Ethanol Self-Administration assessed ethanol consumption. The results showed that the KD blocked the increase in ethanol consumption induced by VSD in females. Moreover, among other changes, the KD increased the expression of the ADORA1 and CNR1 genes, which are associated with mechanisms modulating neurotransmission. Our results point to the KD as a useful tool to increase resilience to social stress in female mice.
Collapse
Affiliation(s)
- Laura Torres-Rubio
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Marina D Reguilón
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| | - Susana Mellado
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - María Pascual
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Marta Rodríguez-Arias
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain
| |
Collapse
|
10
|
Nohesara S, Mostafavi Abdolmaleky H, Thiagalingam S. Substance-Induced Psychiatric Disorders, Epigenetic and Microbiome Alterations, and Potential for Therapeutic Interventions. Brain Sci 2024; 14:769. [PMID: 39199463 PMCID: PMC11352452 DOI: 10.3390/brainsci14080769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 09/01/2024] Open
Abstract
Substance use disorders (SUDs) are complex biopsychosocial diseases that cause neurocognitive deficits and neurological impairments by altering the gene expression in reward-related brain areas. Repeated drug use gives rise to alterations in DNA methylation, histone modifications, and the expression of microRNAs in several brain areas that may be associated with the development of psychotic symptoms. The first section of this review discusses how substance use contributes to the development of psychotic symptoms via epigenetic alterations. Then, we present more evidence about the link between SUDs and brain epigenetic alterations. The next section presents associations between paternal and maternal exposure to substances and epigenetic alterations in the brains of offspring and the role of maternal diet in preventing substance-induced neurological impairments. Then, we introduce potential therapeutic agents/approaches such as methyl-rich diets to modify epigenetic alterations for alleviating psychotic symptoms or depression in SUDs. Next, we discuss how substance use-gut microbiome interactions contribute to the development of neurological impairments through epigenetic alterations and how gut microbiome-derived metabolites may become new therapeutics for normalizing epigenetic aberrations. Finally, we address possible challenges and future perspectives for alleviating psychotic symptoms and depression in patients with SUDs by modulating diets, the epigenome, and gut microbiome.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Mental Health Research Center, Psychosocial Health Research Institute, Department of Psychiatry, School of Medicine, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Hamid Mostafavi Abdolmaleky
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Nutrition/Metabolism Laboratory, Department of Surgery, BIDMC, Harvard Medical School, Boston, MA 02215, USA
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
11
|
Lu Y, George J. Interaction between fatty acid oxidation and ethanol metabolism in liver. Am J Physiol Gastrointest Liver Physiol 2024; 326:G483-G494. [PMID: 38573193 PMCID: PMC11901390 DOI: 10.1152/ajpgi.00281.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/13/2024] [Accepted: 02/26/2024] [Indexed: 04/05/2024]
Abstract
Fatty acid oxidation (FAO) releases the energy stored in fat to maintain basic biological processes. Dehydrogenation is a major way to oxidize fatty acids, which needs NAD+ to accept the released H+ from fatty acids and form NADH, which increases the ratio of NADH/NAD+ and consequently inhibits FAO leading to the deposition of fat in the liver, which is termed fatty liver or steatosis. Consumption of alcohol (ethanol) initiates simple steatosis that progresses to alcoholic steatohepatitis, which constitutes a spectrum of liver disorders called alcohol-associated liver disease (ALD). ALD is linked to ethanol metabolism. Ethanol is metabolized by alcohol dehydrogenase (ADH), microsomal ethanol oxidation system (MEOS), mainly cytochrome P450 2E1 (CYP2E1), and catalase. ADH also requires NAD+ to accept the released H+ from ethanol. Thus, ethanol metabolism by ADH leads to increased ratio of NADH/NAD+, which inhibits FAO and induces steatosis. CYP2E1 directly consumes reducing equivalent NADPH to oxidize ethanol, which generates reactive oxygen species (ROS) that lead to cellular injury. Catalase is mainly present in peroxisomes, where very long-chain fatty acids and branched-chain fatty acids are oxidized, and the resultant short-chain fatty acids will be further oxidized in mitochondria. Peroxisomal FAO generates hydrogen peroxide (H2O2), which is locally decomposed by catalase. When ethanol is present, catalase uses H2O2 to oxidize ethanol. In this review, we introduce FAO (including α-, β-, and ω-oxidation) and ethanol metabolism (by ADH, CYP2E1, and catalase) followed by the interaction between FAO and ethanol metabolism in the liver and its pathophysiological significance.
Collapse
Affiliation(s)
- Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards College of Medicine, Marshall University, Huntington, West Virginia, United States
| | - Joseph George
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, Japan
| |
Collapse
|
12
|
Kumar M, Bhatt B, Gusain C, Mahajan N, Bishnoi M. Sex-specific effects of ketogenic diet on anxiety-like behavior and neuroimmune response in C57Bl/6J mice. J Nutr Biochem 2024; 127:109591. [PMID: 38311044 DOI: 10.1016/j.jnutbio.2024.109591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/06/2024]
Abstract
The ketogenic diet (KD) has been shown to reduce anxiety and enhance cognitive functions in neurological diseases. However, the sex-specific effects of KD on anxiety-like behavior in healthy individuals and the underlying molecular mechanisms contributing to these effects, including neuroinflammation, are unelucidated. This study investigated the sex-specific effects of KD on anxiety-like behavior and the neuroimmune response in the prefrontal cortex (PFC) and hippocampus of healthy C57BL/6J male and female mice. Animals were fed either a control diet (CD- 17% fat, 65% carb, 18% protein) or a KD (80% fat, 5% carb, 15% protein) for 4 weeks. KD increased the levels of circulating β-hydroxybutyrate (BHB) both in males and females. However, PFC BHB levels were found to be elevated only in KD males. Moreover, KD did not affect the behavior of females but improved motor abilities and reduced anxiety levels in males. KD suppressed the mRNA expression of the pan microglial markers (Cd68, P2ry12) and induced morphological changes in the male PFC microglia. A sex-specific decrease in IL1β and an increase in IL-10 levels was found in the PFC of KD males. A similar trend was observed in the hippocampus of males where KD reduced the mRNA expression of P2ry12, Il1β, and cFos. Additionally, BHB increased the production of IL-10 whereas it decreased the production of IL1β from human microglia in in-vitro conditions. In summary, these results demonstrate that the anxiolytic and motor function enhancement abilities of KD are male-specific. Reduced pro-inflammatory and improved anti-inflammatory factors in the male PFC and hippocampus may underlie these effects.
Collapse
Affiliation(s)
- Mohit Kumar
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India; Adjunct faculty, Regional Centre for Biotechnology, Faridabad, Haryana, India.
| | - Babita Bhatt
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Chitralekha Gusain
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Nayan Mahajan
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Food & Nutrition Biotechnology Division, National Agri-Food Biotechnology Institute, S.A.S Nagar, Sector 81 (Knowledge City), Punjab, India
| |
Collapse
|
13
|
Choi AH, Delgado M, Chen KY, Chung ST, Courville A, Turner SA, Yang S, Airaghi K, Dustin I, McGurrin P, Wu T, Hallett M, Ehrlich DJ. A randomized feasibility trial of medium chain triglyceride-supplemented ketogenic diet in people with Parkinson's disease. BMC Neurol 2024; 24:106. [PMID: 38561682 PMCID: PMC10983636 DOI: 10.1186/s12883-024-03603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND A ketogenic diet (KD) may benefit people with neurodegenerative disorders marked by mitochondrial depolarization/insufficiency, including Parkinson's disease (PD). OBJECTIVE Evaluate whether a KD supplemented by medium chain triglyceride (MCT-KD) oil is feasible and acceptable for PD patients. Furthermore, we explored the effects of MCT-KD on blood ketone levels, metabolic parameters, levodopa absorption, mobility, nonmotor symptoms, simple motor and cognitive tests, autonomic function, and resting-state electroencephalography (rsEEG). METHODS A one-week in-hospital, double-blind, randomized, placebo-controlled diet (MCT-KD vs. standard diet (SD)), followed by an at-home two-week open-label extension. The primary outcome was KD feasibility and acceptability. The secondary outcome was the change in Timed Up & Go (TUG) on day 7 of the diet intervention. Additional exploratory outcomes included the N-Back task, Unified Parkinson's Disease Rating Scale, Non-Motor Symptom Scale, and rsEEG connectivity. RESULTS A total of 15/16 subjects completed the study. The mean acceptability was 2.3/3, indicating willingness to continue the KD. Day 7 TUG time was not significantly different between the SD and KD groups. The nonmotor symptom severity score was reduced at the week 3 visit and to a greater extent in the KD group. UPDRS, 3-back, and rsEEG measures were not significantly different between groups. Blood ketosis was attained by day 4 in the KD group and to a greater extent at week 3 than in the SD group. The plasma levodopa metabolites DOPAC and dopamine both showed nonsignificant increasing trends over 3 days in the KD vs. SD groups. CONCLUSIONS An MCT-supplemented KD is feasible and acceptable to PD patients but requires further study to understand its effects on symptoms and disease. TRIAL REGISTRATION Trial Registration Number NCT04584346, registration dates were Oct 14, 2020 - Sept 13, 2022.
Collapse
Affiliation(s)
- Alexander H Choi
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
- Mid-Atlantic Permanente Medical Group, Kaiser Permanente Mid-Atlantic States, Rockville, MD, USA.
| | - Melanie Delgado
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kong Y Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber Courville
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Sara A Turner
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Kayla Airaghi
- NIH Clinical Center Nutrition Department, National Institutes of Health, Bethesda, MD, USA
| | - Irene Dustin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Patrick McGurrin
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Tianxia Wu
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark Hallett
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Debra J Ehrlich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
14
|
Tonetto S, Weikop P, Thomsen M. Nutritional ketosis as treatment for alcohol withdrawal symptoms in female C57BL/6J mice. Sci Rep 2024; 14:5092. [PMID: 38429369 PMCID: PMC10907582 DOI: 10.1038/s41598-024-55310-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 02/22/2024] [Indexed: 03/03/2024] Open
Abstract
Upon both acute and prolonged alcohol intake, the brain undergoes a metabolic shift associated with increased acetate metabolism and reduced glucose metabolism, which persists during abstinence, putatively leading to energy depletion in the brain. This study evaluates the efficacy of ketogenic treatments to rescue psychiatric and neurochemical alterations during long-term alcohol withdrawal. Female mice were intermittently exposed to alcohol vapor or air for three weeks, during which mice were introduced to either a ketogenic diet (KD), control diet supplemented with ketone ester (KE) or remained on control diet (CD). Withdrawal symptoms were assessed over a period of four weeks followed by re-exposure using several behavioral and biochemical tests. Alcohol-exposed mice fed CD displayed long-lasting depressive-like symptoms measured by saccharin preference and tail suspension, as well as decreased norepinephrine levels and serotonin turnover in the hippocampus. Both KD and KE rescued anhedonia for up to three weeks of abstinence. KD mice showed higher latency to first immobility in the tail suspension test, as well as lower plasma cholesterol levels. Our findings show promising effects of nutritional ketosis in ameliorating alcohol withdrawal symptoms in mice. KD seemed to better rescue these symptoms compared to KE.
Collapse
Affiliation(s)
- Simone Tonetto
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark
- Copenhagen Center for Translational Research, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Copenhagen, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - Morgan Thomsen
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Copenhagen, Denmark.
- Copenhagen Center for Translational Research, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Copenhagen, Denmark.
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Laboratory of Neuropsychiatry, Mental Health Center Copenhagen, Copenhagen University Hospital - Mental Health Services CPH, Hovedvejen 17, 1., 2000, Frederiksberg, Denmark.
| |
Collapse
|
15
|
Wiers CE, Manza P, Wang GJ, Volkow ND. Ketogenic diet reduces a neurobiological craving signature in inpatients with alcohol use disorder. Front Nutr 2024; 11:1254341. [PMID: 38410637 PMCID: PMC10895037 DOI: 10.3389/fnut.2024.1254341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
Background and aims Increasing evidence suggests that a ketogenic (high-fat, low-carbohydrate) diet (KD) intervention reduces alcohol withdrawal severity and alcohol craving in individuals with alcohol use disorder (AUD) by shifting brain energetics from glucose to ketones. We hypothesized that the KD would reduce a neurobiological craving signature when individuals undergoing alcohol detoxification treatment were exposed to alcohol cues. Methods We performed a secondary analysis of functional magnetic resonance data of 33 adults with an AUD who were randomized to a KD (n = 19) or a standard American diet (SA; n = 14) and underwent 3 weeks of inpatient alcohol detoxification treatment. Once per week, participants performed an alcohol cue-reactivity paradigm with functional magnetic resonance imaging. We extracted brain responses to food and alcohol cues and quantified the degree to which each set of brain images shared a pattern of activation with a recently established 'Neurobiological Craving Signature' (NCS). We then performed a group-by-time repeated measures ANOVA to test for differences in craving signature expression between the dietary groups over the three-week treatment period. We also correlated these expression patterns with self-reported wanting ratings for alcohol cues. Results For alcohol relative to food cues, there was a main effect of group, such that the KD group showed lower NCS expression across all 3 weeks of treatment. The main effect of time and the group-by-time interaction were not significant. Self-reported wanting for alcohol cues reduced with KD compared to SA but did not correlate with the NCS score. Conclusion A ketogenic diet reduces self-reported alcohol wanting, and induced lower NCS to alcohol cues during inpatient treatment for AUD. However, in the KD group alcohol wanting continued to decrease across the 3 weeks of abstinence while the NCS scores remained stable, suggesting that this cue-induced NCS may not fully capture ongoing, non-cue-induced alcohol desire.
Collapse
Affiliation(s)
- Corinde E. Wiers
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
- Center for Studies of Addiction, Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter Manza
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Gene-Jack Wang
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D. Volkow
- Laboratory of Neuroimaging (LNI), National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| |
Collapse
|
16
|
Okhuarobo A, Kreifeldt M, Gandhi PJ, Lopez C, Martinez B, Fleck K, Bajo M, Bhattacharyya P, Dopico AM, Roberto M, Roberts AJ, Homanics GE, Contet C. Ethanol's interaction with BK channel α subunit residue K361 does not mediate behavioral responses to alcohol in mice. Mol Psychiatry 2024; 29:529-542. [PMID: 38135755 PMCID: PMC11116116 DOI: 10.1038/s41380-023-02346-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
Large conductance potassium (BK) channels are among the most sensitive molecular targets of ethanol and genetic variations in the channel-forming α subunit have been nominally associated with alcohol use disorders. However, whether the action of ethanol at BK α influences the motivation to drink alcohol remains to be determined. To address this question, we first tested the effect of systemically administered BK channel modulators on voluntary alcohol consumption in C57BL/6J males. Penitrem A (blocker) exerted dose-dependent effects on moderate alcohol intake, while paxilline (blocker) and BMS-204352 (opener) were ineffective. Because pharmacological manipulations are inherently limited by non-specific effects, we then sought to investigate the behavioral relevance of ethanol's direct interaction with BK α by introducing in the mouse genome a point mutation known to render BK channels insensitive to ethanol while preserving their physiological function. The BK α K361N substitution prevented ethanol from reducing spike threshold in medial habenula neurons. However, it did not alter acute responses to ethanol in vivo, including ataxia, sedation, hypothermia, analgesia, and conditioned place preference. Furthermore, the mutation did not have reproducible effects on alcohol consumption in limited, continuous, or intermittent access home cage two-bottle choice paradigms conducted in both males and females. Notably, in contrast to previous observations made in mice missing BK channel auxiliary β subunits, the BK α K361N substitution had no significant impact on ethanol intake escalation induced by chronic intermittent alcohol vapor inhalation. It also did not affect the metabolic and locomotor consequences of chronic alcohol exposure. Altogether, these data suggest that the direct interaction of ethanol with BK α does not mediate the alcohol-related phenotypes examined here in mice.
Collapse
Affiliation(s)
- Agbonlahor Okhuarobo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Max Kreifeldt
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Pauravi J Gandhi
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Catherine Lopez
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Briana Martinez
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Kiera Fleck
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Michal Bajo
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | | | - Alex M Dopico
- University of Tennessee Health Science Center, Department of Pharmacology, Addiction Science, and Toxicology, Memphis, TN, USA
| | - Marisa Roberto
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Amanda J Roberts
- The Scripps Research Institute, Animals Models Core Facility, La Jolla, CA, USA
| | - Gregg E Homanics
- University of Pittsburgh, Department of Anesthesiology and Perioperative Medicine, Pittsburgh, PA, USA
| | - Candice Contet
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA.
| |
Collapse
|
17
|
Li X, Shi Z, Todaro DR, Pond T, Byanyima JI, Vesslee SA, Reddy R, Nanga RPR, Kass G, Ramchandani V, Kranzler HR, Vendruscolo JCM, Vendruscolo LF, Wiers CE. Ketone Supplementation Dampens Subjective and Objective Responses to Alcohol: Evidence From a Preclinical Rat Study and a Randomized, Cross-Over Trial in Healthy Volunteers. Int J Neuropsychopharmacol 2024; 27:pyae009. [PMID: 38315678 PMCID: PMC10901540 DOI: 10.1093/ijnp/pyae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Previous preclinical and human studies have shown that a high-fat ketogenic diet and ketone supplements (KS) are efficacious in reducing alcohol craving, alcohol consumption, and signs of alcohol withdrawal. However, the effects of KS on alcohol sensitivity are unknown. METHODS In this single-blind, cross-over study, 10 healthy participants (3 females) were administered a single, oral dose of a KS (25 g of ketones from D-β-hydroxybutyric acid and R-1,3-butanediol) or placebo 30 minutes before an oral alcohol dose (0.25 g/kg for women; 0.31 g/kg for men). Assessments of breath alcohol concentration and blood alcohol levels (BAL) and responses on the Drug Effect Questionnaire were repeatedly obtained over 180 minutes after alcohol consumption. In a parallel preclinical study, 8 Wistar rats (4 females) received an oral gavage of KS (0.42 g ketones/kg), water, or the sweetener allulose (0.58 g/kg) followed 15 minutes later by an oral alcohol dose (0.8 g/kg). BAL was monitored for 240 minutes after alcohol exposure. RESULTS In humans, the intake of KS before alcohol significantly blunted breath alcohol concentration and BAL, reduced ratings of alcohol liking and wanting more, and increased disliking for alcohol. In rats, KS reduced BAL more than either allulose or water. CONCLUSION KS altered physiological and subjective responses to alcohol in both humans and rats, and the effects were likely not mediated by the sweetener allulose present in the KS drink. Therefore, KS could potentially reduce the intoxicating effects of alcohol.
Collapse
Affiliation(s)
- Xinyi Li
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Zhenhao Shi
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Dustin R Todaro
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Timothy Pond
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Juliana I Byanyima
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Sianneh A Vesslee
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rishika Reddy
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ravi Prakash Reddy Nanga
- University of Pennsylvania Perelman School of Medicine, Department of Radiology, Philadelphia, Pennsylvania, USA
| | - Gabriel Kass
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Vijay Ramchandani
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA
| | - Henry R Kranzler
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Leandro F Vendruscolo
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, USA
| | - Corinde E Wiers
- Center for Studies of Addiction, Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
White B, Sirohi S. A Complex Interplay between Nutrition and Alcohol use Disorder: Implications for Breaking the Vicious Cycle. Curr Pharm Des 2024; 30:1822-1837. [PMID: 38797900 DOI: 10.2174/0113816128292367240510111746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
Approximately 16.5% of the United States population met the diagnostic criteria for substance use disorder (SUD) in 2021, including 29.5 million individuals with alcohol use disorder (AUD). Individuals with AUD are at increased risk for malnutrition, and impairments in nutritional status in chronic alcohol users can be detrimental to physical and emotional well-being. Furthermore, these nutritional deficiencies could contribute to the never-ending cycle of alcoholism and related pathologies, thereby jeopardizing the prospects of recovery and treatment outcomes. Improving nutritional status in AUD patients may not only compensate for general malnutrition but could also reduce adverse symptoms during recovery, thereby promoting abstinence and successful treatment of AUD. In this review, we briefly summarize alterations in the nutritional status of people with addictive disorders, in addition to the underlying neurobiological mechanisms and clinical implications regarding the role of nutritional intervention in recovery from alcohol use disorder.
Collapse
Affiliation(s)
- Brooke White
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Sunil Sirohi
- Laboratory of Endocrine and Neuropsychiatric Disorders, Division of Basic Pharmaceutical Sciences, College of Pharmacy, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
19
|
Lu JF, Zhu MQ, Xia B, Zhang NN, Liu XP, Liu H, Zhang RX, Xiao JY, Yang H, Zhang YQ, Li XM, Wu JW. GDF15 is a major determinant of ketogenic diet-induced weight loss. Cell Metab 2023; 35:2165-2182.e7. [PMID: 38056430 DOI: 10.1016/j.cmet.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/27/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
A ketogenic diet (KD) has been promoted as an obesity management diet, yet its underlying mechanism remains elusive. Here we show that KD reduces energy intake and body weight in humans, pigs, and mice, accompanied by elevated circulating growth differentiation factor 15 (GDF15). In GDF15- or its receptor GFRAL-deficient mice, these effects of KD disappeared, demonstrating an essential role of GDF15-GFRAL signaling in KD-mediated weight loss. Gdf15 mRNA level increases in hepatocytes upon KD feeding, and knockdown of Gdf15 by AAV8 abrogated the obesity management effect of KD in mice, corroborating a hepatic origin of GDF15 production. We show that KD activates hepatic PPARγ, which directly binds to the regulatory region of Gdf15, increasing its transcription and production. Hepatic Pparγ-knockout mice show low levels of plasma GDF15 and significantly diminished obesity management effects of KD, which could be restored by either hepatic Gdf15 overexpression or recombinant GDF15 administration. Collectively, our study reveals a previously unexplored GDF15-dependent mechanism underlying KD-mediated obesity management.
Collapse
Affiliation(s)
- Jun Feng Lu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Meng Qing Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Bo Xia
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Na Na Zhang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao Peng Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Huan Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Rui Xin Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jun Ying Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hui Yang
- National Health Commission (NHC) Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Ying Qi Zhang
- State Key Laboratory of Cancer Biology, Biotechnology Center, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao Miao Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, China.
| | - Jiang Wei Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
20
|
Wiers CE, Manza P, Wang GJ, Volkow ND. Ketogenic diet reduces a neurobiological craving signature in alcohol use disorder. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.25.23296094. [PMID: 37886532 PMCID: PMC10602038 DOI: 10.1101/2023.09.25.23296094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Background and Aims Increasing evidence suggests that a ketogenic (high-fat, low-carbohydrate) diet intervention reduces alcohol withdrawal severity and alcohol craving in individuals with alcohol use disorder (AUD) by shifting brain energetics from glucose to ketones. We hypothesized that the ketogenic diet would reduce a brain craving signature when individuals undergoing alcohol detoxification treatment were exposed to alcohol cues. Methods We performed a secondary analysis of functional magnetic resonance data of n=33 adults with an AUD were randomized to a ketogenic diet (n=19) or a standard American diet (n=14) and underwent three weeks of inpatient alcohol detoxification treatment. Once per week, participants performed an alcohol cue-reactivity paradigm with functional magnetic resonance imaging. We extracted brain responses to food and alcohol cues and quantified the degree to which each set of brain images shared a pattern of activation with a recently validated 'Neurobiological Craving Signature' (NCS). We then performed a group-by-time repeated measures ANOVA to test for differences in craving signature expression between the dietary groups over the three-week treatment period. We also correlated these expression patterns with self-reported wanting ratings for alcohol cues. Results For alcohol relative to food cues, there was a main effect of group, such that the ketogenic diet group showed lower NCS expression across all three weeks of treatment. The main effect of time and the group-by-time interaction were not significant. Self-reported wanting for alcohol cues reduced with KD compared to SA but did not correlate with the NCS score. Conclusions A ketogenic diet reduces self-reported alcohol wanting, and induced lower brain craving signatures to alcohol cues during inpatient treatment for AUD.
Collapse
|
21
|
Li X, Shi Z, Todaro D, Pond T, Byanyima J, Vesslee S, Reddy R, Reddy Nanga RP, Kass G, Ramchandani V, Kranzler HR, Vendruscolo JCM, Vendruscolo LF, Wiers CE. Ketone supplementation dampens subjective and objective responses to alcohol in rats and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.23.558269. [PMID: 37790364 PMCID: PMC10542198 DOI: 10.1101/2023.09.23.558269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Previous preclinical and human studies have shown that high-fat ketogenic diet and ketone supplements (KS) are efficacious in reducing alcohol craving, alcohol consumption, and signs of alcohol withdrawal. However, the effects of KS on alcohol sensitivity are unknown. In this single-blind, cross-over study, 10 healthy participants (3 females) were administered a single, oral dose of a KS (25 g of ketones from D-β-hydroxybutyric acid and R-1,3-butanediol) or placebo 30 min prior to an oral alcohol dose (0.25 g/kg for women; 0.31 g/kg for men). Assessments of breath alcohol concentration (BrAC) and blood alcohol levels (BAL) and responses on the Drug Effect Questionnaire were repeatedly obtained over 180 min after alcohol consumption. In a parallel preclinical study, 8 Wistar rats (4 females) received an oral gavage of KS (0.42 g ketones/kg), water, or the sweetener allulose (0.58 g/kg) followed 15 min later by an oral alcohol dose (0.8 g/kg). BAL were monitored for 240 min after alcohol exposure. In humans, the intake of KS prior to alcohol significantly blunted BrAC and BAL, reduced ratings of alcohol liking and wanting, and increased disliking for alcohol. In rats, KS reduced BAL more than either allulose or water. In conclusion, KS altered physiological and subjective responses to alcohol in both humans and rats and the effects were likely not mediated by the sweetener allulose present in the KS drink. Therefore, KS could potentially reduce the intoxicating and rewarding effects of alcohol and thus be a novel intervention for treating alcohol use disorder.
Collapse
|
22
|
Li K, Wang WH, Wu JB, Xiao WH. β-hydroxybutyrate: A crucial therapeutic target for diverse liver diseases. Biomed Pharmacother 2023; 165:115191. [PMID: 37487440 DOI: 10.1016/j.biopha.2023.115191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023] Open
Abstract
β-hydroxybutyrate (β-HB), the most abundant ketone body, is produced primarily in the liver and acts as a substitute energy fuel to provide energy to extrahepatic tissues in the event of hypoglycemia or glycogen depletion. We now have an improved understanding of β-HB as a signal molecule and epigenetic regulatory factor as a result of intensive research over the last ten years. Because β-HB regulates various physiological and pathological processes, it may have a potential role in the treatment of metabolic diseases. The liver is the most significant metabolic organ, and the part that β-HB plays in liver disorders is receiving increasing attention. In this review, we summarize the therapeutic effects of β-HB on liver diseases and its underlying mechanisms of action. Moreover, we explore the prospects of exogenous supplements and endogenous ketosis including fasting, caloric restriction (CR), ketogenic diet (KD), and exercise as adjuvant nutritional therapies to protect the liver from damage and provide insights and strategies for exploring the treatment of various liver diseases.
Collapse
Affiliation(s)
- Ke Li
- Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Wen-Hong Wang
- Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Jia-Bin Wu
- Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Wei-Hua Xiao
- Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai 200438, China; Shanghai Key Lab of Human Performance, Shanghai University of Sport, Shanghai 200438, China.
| |
Collapse
|
23
|
Kong D, Sun JX, Yang JQ, Li YS, Bi K, Zhang ZY, Wang KH, Luo HY, Zhu M, Xu Y. Ketogenic diet: a potential adjunctive treatment for substance use disorders. Front Nutr 2023; 10:1191903. [PMID: 37575322 PMCID: PMC10414993 DOI: 10.3389/fnut.2023.1191903] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Substance use disorders (SUD) can lead to serious health problems, and there is a great interest in developing new treatment methods to alleviate the impact of substance abuse. In recent years, the ketogenic diet (KD) has shown therapeutic benefits as a dietary therapy in a variety of neurological disorders. Recent studies suggest that KD can compensate for the glucose metabolism disorders caused by alcohol use disorder by increasing ketone metabolism, thereby reducing withdrawal symptoms and indicating the therapeutic potential of KD in SUD. Additionally, SUD often accompanies increased sugar intake, involving neural circuits and altered neuroplasticity similar to substance addiction, which may induce cross-sensitization and increased use of other abused substances. Reducing carbohydrate intake through KD may have a positive effect on this. Finally, SUD is often associated with mitochondrial damage, oxidative stress, inflammation, glia dysfunction, and gut microbial disorders, while KD may potentially reverse these abnormalities and serve a therapeutic role. Although there is much indirect evidence that KD has a positive effect on SUD, the small number of relevant studies and the fact that KD leads to side effects such as metabolic abnormalities, increased risk of malnutrition and gastrointestinal symptoms have led to the limitation of KD in the treatment of SUD. Here, we described the organismal disorders caused by SUD and the possible positive effects of KD, aiming to provide potential therapeutic directions for SUD.
Collapse
Affiliation(s)
- Deshenyue Kong
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-xue Sun
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji-qun Yang
- Third People’s Hospital of Kunming City/Drug Rehabilitation Hospital of Kunming City, Kunming, China
| | - Yuan-sen Li
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ke Bi
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zun-yue Zhang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Kun-hua Wang
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
| | - Hua-you Luo
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Mei Zhu
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yu Xu
- General Hospital of Eastern Theater Command, Nanjing, China
- Yunnan Technological Innovation Centre of Drug Addiction Medicine, Yunnan University, Kunming, China
- First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
24
|
Soni ND, Swain A, Jacobs P, Juul H, Armbruster R, Nanga RPR, Nath K, Wiers C, Detre J, Reddy R. In vivo assessment of β-hydroxybutyrate metabolism in mouse brain using deuterium ( 2 H) MRS. Magn Reson Med 2023; 90:259-269. [PMID: 36971349 PMCID: PMC10662955 DOI: 10.1002/mrm.29648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 05/01/2023]
Abstract
PURPOSE To monitor the metabolic turnover of β-hydroxybutyrate (BHB) oxidation using 2 H-MRS in conjunction with intravenous administration of 2 H labeled BHB. METHODS Nine-month-old mice were infused with [3,4,4,4]-2 H4 -BHB (d4 -BHB; 3.11 g/kg) through the tail vein using a bolus variable infusion rate for a period of 90 min. The labeling of downstream cerebral metabolites from the oxidative metabolism of d4 -BHB was monitored using 2 H-MRS spectra acquired with a home-built 2 H surface coil on a 9.4T preclinical MR scanner with a temporal resolution of 6.25 min. An exponential model was fit to the BHB and glutamate/glutamine (Glx) turnover curves to determine rate constants of metabolite turnover and to aid in the visualization of metabolite time courses. RESULTS Deuterium label was incorporated into Glx from BHB metabolism through the tricarboxylic acid (TCA) cycle, with an increase in the level of [4,4]-2 H2 -Glx (d2 -Glx) over time and reaching a quasi-steady state concentration of ∼0.6 ± 0.1 mM following 30 min of infusion. Complete oxidative metabolic breakdown of d4 -BHB also resulted in the formation of semi-heavy water (HDO), with a four-fold (10.1 to ∼42.1 ± 7.3 mM) linear (R2 = 0.998) increase in its concentration by the end of infusion. The rate constant of Glx turnover from d4 -BHB metabolism was determined to be 0.034 ± 0.004 min-1 . CONCLUSION 2 H-MRS can be used to monitor the cerebral metabolism of BHB with its deuterated form by measuring the downstream labeling of Glx. The integration of 2 H-MRS with deuterated BHB substrate provides an alternative and clinically promising MRS tool to detect neurometabolic fluxes in healthy and disease conditions.
Collapse
Affiliation(s)
- Narayan Datt Soni
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anshuman Swain
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA
| | - Paul Jacobs
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA
| | - Halvor Juul
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Armbruster
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ravi Prakash Reddy Nanga
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kavindra Nath
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Corinde Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - John Detre
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Ravinder Reddy
- Department of Radiology, Center for Advanced Metabolic Imaging in Precision Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Trinko R, Diaz DM, Foscue E, Thompson SL, Taylor JR, DiLeone RJ. Ketogenic diet enhances the effects of oxycodone in mice. Sci Rep 2023; 13:7507. [PMID: 37160959 PMCID: PMC10170077 DOI: 10.1038/s41598-023-33458-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
Opioids have been used to manage pain for thousands of years, but they have significant potential for abuse. Prescription opioids, like oxycodone, are associated with 32% of overdoses, that have reached a total of 75,673 deaths in 2021. A major challenge is maximizing their therapeutic potential while minimizing the negative side effects including opioid use disorder (OUD). The Ketogenic Diet (KD) has been reported to reduce pain and decrease the severity of alcohol use disorder, yet its effects on oxycodone responses remain unknown. KD mice displayed increased oxycodone-induced locomotor activity and enhanced antinociceptive effects of oxycodone, suggesting a dietary effect on opiate sensitivity. Male KD mice exposed to chronic oxycodone exhibited increased naloxone-induced jumps, suggesting a sex-specific effect of diet on opioid withdrawal. Consistent with this, male KD mice self-administered less oxycodone while female KD mice did not differ from controls. Finally, no effect of KD on motivation to obtain oxycodone was observed during a progressive ratio schedule. These data suggest sex-biased effects of KD on responses to opioids that should be considered and potentially leveraged in both clinical pain management and treatment of OUD.
Collapse
Affiliation(s)
- R Trinko
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - D M Diaz
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - E Foscue
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - S L Thompson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - J R Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA
| | - R J DiLeone
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06519, USA.
| |
Collapse
|
26
|
Willemse L, Terburgh K, Louw R. A ketogenic diet alters mTOR activity, systemic metabolism and potentially prevents collagen degradation associated with chronic alcohol consumption in mice. Metabolomics 2023; 19:43. [PMID: 37076659 PMCID: PMC10115735 DOI: 10.1007/s11306-023-02006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION A ketogenic diet (KD), which is a high fat, low carbohydrate diet has been shown to inhibit the mammalian target of rapamycin (mTOR) pathway and alter the redox state. Inhibition of the mTOR complex has been associated with the attenuation and alleviation of various metabolic and- inflammatory diseases such as neurodegeneration, diabetes, and metabolic syndrome. Various metabolic pathways and signalling mechanisms have been explored to assess the therapeutic potential of mTOR inhibition. However, chronic alcohol consumption has also been reported to alter mTOR activity, the cellular redox- and inflammatory state. Thus, a relevant question that remains is what effect chronic alcohol consumption would have on mTOR activity and overall metabolism during a KD-based intervention. OBJECTIVES The aim of this study was to evaluate the effect of alcohol and a KD on the phosphorylation of the mTORC1 target p70S6K, systemic metabolism as well as the redox- and inflammatory state in a mouse model. METHODS Mice were fed either a control diet with/without alcohol or a KD with/without alcohol for three weeks. After the dietary intervention, samples were collected and subjected towards western blot analysis, multi-platform metabolomics analysis and flow cytometry. RESULTS Mice fed a KD exhibited significant mTOR inhibition and reduction in growth rate. Alcohol consumption alone did not markedly alter mTOR activity or growth rate but moderately increased mTOR inhibition in mice fed a KD. In addition, metabolic profiling showed alteration of several metabolic pathways as well as the redox state following consumption of a KD and alcohol. A KD was also observed to potentially prevent bone loss and collagen degradation associated with chronic alcohol consumption, as indicated by hydroxyproline metabolism. CONCLUSION This study sheds light on the influence that a KD alongside alcohol intake can exert on not just mTOR, but also their effect on metabolic reprogramming and the redox state.
Collapse
Affiliation(s)
- Luciano Willemse
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
27
|
Jerlhag E. The therapeutic potential of glucagon-like peptide-1 for persons with addictions based on findings from preclinical and clinical studies. Front Pharmacol 2023; 14:1063033. [PMID: 37063267 PMCID: PMC10097922 DOI: 10.3389/fphar.2023.1063033] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Although the multifaceted mechanisms underlying alcohol use disorder (AUD) have been partially defined, the neurobiological complexity of this disorder is yet to be unraveled. One of the systems that have gained attention in recent times is the gut–brain axis. Although numerous peptides participate in this axis, glucagon-like peptide-1 (GLP-1) plays a central role. GLP-1 is a crucial anorexigenic peptide, with potent abilities to reduce food intake and body weight. The physiological complexity of GLP-1 entails glucose homeostasis, gastrointestinal motility, and the release of insulin and glucagon. As reviewed in this study, acute or repeated treatment with GLP-1 receptor (GLP-1R) agonists decreases alcohol consumption in rodents. Moreover, the abilities of alcohol to promote hyperlocomotion, dopamine release in the nucleus accumbens, and reward in the conditioned place preference paradigm are all suppressed by GLP-1R ligands. Moreover, activation of GLP-1R suppresses the motivation to consume alcohol, alcohol-seeking behaviors, and relapse drinking in male rodents. Similarly, abstinence symptoms experienced during alcohol withdrawal are attenuated by activation of the GLP-1 pathway. On a similar note, the activation of GLP-1 receptors within areas of the brain that are processing reward modulates these alcohol-related responses. Another area that is crucial for this ability is the nucleus of the solitary tract, which is where GLP-1 is produced and from which GLP-1-containing neurons project to areas of reward. These findings may have clinical relevance as AUD is associated with polymorphisms in GLP-1-related genes. Although a GLP-1R agonist does not alter alcohol intake in AUD patients, it reduces this consumption in a sub-population of obese AUD individuals. Given the uncertainty of this outcome, additional clinical studies of obese AUD patients should explore the effects of the GLP-1R agonists on alcohol intake and body weight. Furthermore, GLP-1 receptors modulate the behavioral and neurochemical responses to addictive drugs. Taken together, these preclinical and clinical findings imply that the GLP-1 pathway plays a role in the complex mechanisms regulating alcohol and drug consumption patterns, unveiling a novel aspect of addiction medicine.
Collapse
|
28
|
Fitzpatrick-Schmidt T, Edwards S. Cortisol as a risk biomarker to guide recovery from substance use disorders. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2023; 47:435-437. [PMID: 36811294 PMCID: PMC10050113 DOI: 10.1111/acer.15018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/30/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023]
Affiliation(s)
| | - Scott Edwards
- Department of Physiology and Comprehensive Alcohol-HIV/AIDS Research Center, LSU Health-New Orleans
| |
Collapse
|
29
|
Bach P, de Timary P, Gründer G, Cumming P. Molecular Imaging Studies of Alcohol Use Disorder. Curr Top Behav Neurosci 2023. [PMID: 36639552 DOI: 10.1007/7854_2022_414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Alcohol use disorder (AUD) is a serious public health problem in many countries, bringing a gamut of health risks and impairments to individuals and a great burden to society. Despite the prevalence of a disease model of AUD, the current pharmacopeia does not present reliable treatments for AUD; approved treatments are confined to a narrow spectrum of medications engaging inhibitory γ-aminobutyric acid (GABA) neurotransmission and possibly excitatory N-methyl-D-aspartate (NMDA) receptors, and opioid receptor antagonists. Molecular imaging with positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can open a window into the living brain and has provided diverse insights into the pathology of AUD. In this narrative review, we summarize the state of molecular imaging findings on the pharmacological action of ethanol and the neuropathological changes associated with AUD. Laboratory and preclinical imaging results highlight the interactions between ethanol and GABA A-type receptors (GABAAR), but the interpretation of such results is complicated by subtype specificity. An abundance of studies with the glucose metabolism tracer fluorodeoxyglucose (FDG) concur in showing cerebral hypometabolism after ethanol challenge, but there is relatively little data on long-term changes in AUD. Alcohol toxicity evokes neuroinflammation, which can be tracked using PET with ligands for the microglial marker translocator protein (TSPO). Several PET studies show reversible increases in TSPO binding in AUD individuals, and preclinical results suggest that opioid-antagonists can rescue from these inflammatory responses. There are numerous PET/SPECT studies showing changes in dopaminergic markers, generally consistent with an impairment in dopamine synthesis and release among AUD patients, as seen in a number of other addictions; this may reflect the composite of an underlying deficiency in reward mechanisms that predisposes to AUD, in conjunction with acquired alterations in dopamine signaling. There is little evidence for altered serotonin markers in AUD, but studies with opioid receptor ligands suggest a specific up-regulation of the μ-opioid receptor subtype. Considerable heterogeneity in drinking patterns, gender differences, and the variable contributions of genetics and pre-existing vulnerability traits present great challenges for charting the landscape of molecular imaging in AUD.
Collapse
Affiliation(s)
- Patrick Bach
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.
| | - Philippe de Timary
- Department of Adult Psychiatry, Cliniques Universitaires Saint-Luc and Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Gerhard Gründer
- Department of Molecular Neuroimaging, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
- International Centre for Education and Research in Neuropsychiatry (ICERN), Samara State Medical University, Samara, Russia
| |
Collapse
|
30
|
Goutaudier R, Joly F, Mallet D, Bartolomucci M, Guicherd D, Carcenac C, Vossier F, Dufourd T, Boulet S, Deransart C, Chovelon B, Carnicella S. Hypodopaminergic state of the nigrostriatal pathway drives compulsive alcohol use. Mol Psychiatry 2023; 28:463-474. [PMID: 36376463 PMCID: PMC9812783 DOI: 10.1038/s41380-022-01848-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 10/18/2022] [Indexed: 11/16/2022]
Abstract
The neurobiological mechanisms underlying compulsive alcohol use, a cardinal feature of alcohol use disorder, remain elusive. The key modulator of motivational processes, dopamine (DA), is suspected to play an important role in this pathology, but its exact role remains to be determined. Here, we found that rats expressing compulsive-like alcohol use, operationalized as punishment-resistant self-administration, showed a decrease in DA levels restricted to the dorsolateral territories of the striatum, the main output structure of the nigrostriatal DA pathway. We then causally demonstrated that chemogenetic-induced selective hypodopaminergia of this pathway resulted in compulsive-like alcohol self-administration in otherwise resilient rats, accompanied by the emergence of alcohol withdrawal-like motivational impairments (i.e., impaired motivation for a natural reinforcer). Finally, the use of the monoamine stabilizer OSU6162, previously reported to correct hypodopaminergic states, transiently decreased compulsive-like alcohol self-administration in vulnerable rats. These results suggest a potential critical role of tonic nigrostriatal hypodopaminergic states in alcohol addiction and provide new insights into our understanding of the neurobiological mechanisms underlying compulsive alcohol use.
Collapse
Affiliation(s)
- Raphaël Goutaudier
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Fanny Joly
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - David Mallet
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Magali Bartolomucci
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Denis Guicherd
- grid.410529.b0000 0001 0792 4829Service de Biochimie, Biologie Moléculaire, Toxicologie Environnementale, CHU de Grenoble-Alpes Site Nord − Institut de Biologie et de Pathologie, F-38041 Grenoble, France
| | - Carole Carcenac
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Frédérique Vossier
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Thibault Dufourd
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Sabrina Boulet
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Colin Deransart
- grid.462307.40000 0004 0429 3736Inserm, U1216, Univ. Grenoble Alpes, CHU Grenoble Alpes, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Benoit Chovelon
- grid.410529.b0000 0001 0792 4829Service de Biochimie, Biologie Moléculaire, Toxicologie Environnementale, CHU de Grenoble-Alpes Site Nord − Institut de Biologie et de Pathologie, F-38041 Grenoble, France ,grid.4444.00000 0001 2112 9282Univ. Grenoble Alpes, CNRS, DPM, 38000 Grenoble, France
| | - Sebastien Carnicella
- Inserm, U1216, Univ. Grenoble Alpes, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
31
|
Li Z, Vidjro OE, Guo G, Du Y, Zhou Y, Xie Q, Li J, Gao K, Zhou L, Ma T. NLRP3 deficiency decreases alcohol intake controlling anxiety-like behavior via modification of glutamatergic transmission in corticostriatal circuits. J Neuroinflammation 2022; 19:308. [PMID: 36539796 PMCID: PMC9764485 DOI: 10.1186/s12974-022-02666-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Alcohol use disorders result from repeated binge and chronic alcohol consumption followed by negative effects, such as anxiety, upon cessation. This process is associated with the activation of NLRP3 inflammasome-mediated responses. However, whether and how inhibition of the NLRP3 inflammasome alters alcohol intake and anxiety behavior remains unclear. METHODS A combination of drinking-in-the-dark and gavage was established in NLRP3-knockout and control mice. Behavior was assessed by open-field and elevated plus maze tests. Binge alcohol drinking was measured at 2 h and 4 h. A 2 h/4 h/24 h voluntary drinking was determined by a two-bottle choice paradigm. Western blotting and ELISA were applied to examine the levels of the NLRP3 inflammasome and- inflammatory factors, such as IL-1β and TNF-α. Nissl staining was used to measure neuronal injury. The electrophysiological method was used to determine glutamatergic transmission in corticostriatal circuits. In vivo optogenetic LTP and LTD were applied to control the function of corticostriatal circuits on the behavior of mice. MCC950 was used to antagonize the NLRP3 inflammasome. RESULTS The binge alcohol intake was decreased in NLRP3 KO mice compared to the control mice. During alcohol withdrawal, NLRP3 deficiency attenuated anxiety-like behavior and neuronal injury in the mPFC and striatum. Moreover, we discovered that glutamatergic transmission to striatal neurons was reduced in NLRP3 KO mice. Importantly, in vivo optogenetic induction of long-term potentiation (LTP) of corticostriatal circuits reversed the effects of NLRP3 deficiency on glutamatergic transmission and anxiety behavior. We also demonstrated that optogenetic induction of LTD decreased anxiety-like behavior and caused a reduction in glutamatergic transmission. Interestingly, NLRP3 deficiency or inhibition (MCC950 injection) attenuated the anxiety-like behavior, but it did not prevent DID + gavage paradigm-induced a persistent enhancement of drinking in a two-bottle choice at 2 and 4 days into withdrawal. CONCLUSION Our results demonstrate that NLRP3 deficiency decreases binge alcohol intake and anxiety-like behavior through downregulation of glutamatergic transmission in corticostriatal circuits, which may provide an anti-inflammatory target for treating alcohol use disorders.
Collapse
Affiliation(s)
- Ziyi Li
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Olivia Ewi Vidjro
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Gengni Guo
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China ,grid.89957.3a0000 0000 9255 8984Grade 2020 in Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu China
| | - Yanfeng Du
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Yao Zhou
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Qian Xie
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Jiaxin Li
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Keqiang Gao
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| | - Li Zhou
- grid.459791.70000 0004 1757 7869Department of Anesthesiology, Women’s Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, 123 Tianfei Lane, Mochou Road, Nanjing, 210004 Jiangsu China ,grid.89957.3a0000 0000 9255 8984Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu China
| | - Tengfei Ma
- grid.89957.3a0000 0000 9255 8984Institute for Stem Cell and Neural Regeneration and Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, 101 Longmian Street, Nanjing, 211166 Jiangsu China
| |
Collapse
|
32
|
Li X, Shi Z, Byanyima J, Morgan PT, van der Veen JW, Zhang R, Deneke E, Wang GJ, Volkow ND, Wiers CE. Brain glutamate and sleep efficiency associations following a ketogenic diet intervention in individuals with Alcohol Use Disorder. DRUG AND ALCOHOL DEPENDENCE REPORTS 2022; 5:100092. [PMID: 36311277 PMCID: PMC9601174 DOI: 10.1016/j.dadr.2022.100092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background We previously showed that ketogenic diet (KD) was effective in curbing alcohol withdrawal and craving in individuals with alcohol use disorder (AUD). We hypothesized that the clinical benefits were due to improvements in sleep. To test this, we performed a secondary analysis on the KD trial data to (1) examine the effects of KD on total sleep time (TST) and sleep quality and (2) investigate the association between KD-induced alterations in cingulate glutamate concentration and changes in TST and sleep quality. Methods AUD individuals undergoing alcohol detoxification were randomized to receive KD (n=19) or standard American diet (SA; n=14) for three weeks. TST was measured weekly by self-report, GENEActive sleep accelerometer, and X4 Sleep Profiler ambulatory device. Sleep quality was assessed using subjectively ratings of sleep depth and restedness and Sleep Profiler (Sleep Efficiency [%]). Weekly 1H magnetic resonance spectroscopy scans measured cingulate glutamate levels. Results TST was lower in KD than SA and increased with effect of time. Sleep depth, restedness, and Sleep Efficiency improved with time, but exhibited no effect of diet. In KD and SA combined, week 1 cingulate glutamate levels correlated positively with Sleep Efficiency, but not with TST. Conclusions Although cingulate glutamate levels correlated positively with Sleep Efficiency in week 1, KD-induced glutamate elevation did not produce significant sleep improvements. Rather, KD was associated with lower TST than SA. Given the well-established associations between sleep and alcohol relapse, longer follow up assessment of KD's impact on sleep in AUD is warranted.
Collapse
Affiliation(s)
- Xinyi Li
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Zhenhao Shi
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Juliana Byanyima
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Peter T. Morgan
- Department of Psychiatry, Yale University, New Haven, CT, 06519, USA
- Department of Psychiatry, Bridgeport Hospital, Bridgeport, CT, 06610, USA
| | | | - Rui Zhang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Erin Deneke
- Caron Treatment Centers, 243N Galen Hall Rd, Wernersville, PA, 19565, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Nora D. Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
| | - Corinde E. Wiers
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, 20892, USA
- Corresponding author at: 3535 Market St Ste 500, Philadelphia, PA 19104.
| |
Collapse
|
33
|
Li H, Wan X, Wu Z, Zhou Y, Chen R, Xu W, Zhang J, Yang Z, Bai L, Zhang J, Qin F, Wang L, Chen Y, Jiang L, He Y, Wang X, Wei Q, Li S, Dai Y, Chen Y, Wang Y, Wang H, Tian J, Zhao Y, Cen X. β-hydroxybutyrate reduces reinstatement of cocaine conditioned place preference through hippocampal CaMKII-α β-hydroxybutyrylation. Cell Rep 2022; 41:111724. [PMID: 36450263 DOI: 10.1016/j.celrep.2022.111724] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/29/2022] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
Studies have shown the therapeutic effects of a ketogenic diet (KD) on epilepsy, but the effect of a KD on drug reinstatement is largely unclear. This study aims to investigate whether KD consumption possesses therapeutic potential for cocaine reinstatement and the molecular mechanism. We find that a KD significantly reduces cocaine-induced reinstatement in mice, which is accompanied by a markedly elevated level of β-hydroxybutyrate (β-OHB), the most abundant ketone body, in the hippocampus. The underlying mechanism is that β-OHB posttranslationally modifies CaMKII-α with β-hydroxybutyrylation, resulting in significant inhibition of T286 autophosphorylation and downregulation of CaMKII activity. Collectively, our results reveal that β-hydroxybutyrylation is a posttranslational modification of CaMKII-α that plays a critical role in mediating the effect of KD consumption in reducing cocaine reinstatement.
Collapse
Affiliation(s)
- Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuemei Wan
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhixiang Wu
- Faculty of Environmental and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Yuanyi Zhou
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rong Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Xu
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Shenzhen Key Laboratory of Drug Addiction, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen 518055, China
| | - Jiamei Zhang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhen Yang
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Bai
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhang
- Histology and Imaging Platform, Core Facilities of West China Hospital, Sichuan University, Chengdu 610041, China
| | - Feng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaxing Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linhong Jiang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuman He
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaojie Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qingfan Wei
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shu Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanping Dai
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuanyuan Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yonghai Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Hongbo Wang
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Jingwei Tian
- Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China
| | - Yinglan Zhao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
34
|
Suk FM, Wu CY, Chiu WC, Chien CY, Chen TL, Liao YJ. HMGCS2 Mediation of Ketone Levels Affects Sorafenib Treatment Efficacy in Liver Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27228015. [PMID: 36432116 PMCID: PMC9697984 DOI: 10.3390/molecules27228015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Primary liver cancer is the fifth leading death of cancers in men, and hepatocellular carcinoma (HCC) accounts for approximately 90% of all primary liver cancer cases. Sorafenib is a first-line drug for advanced-stage HCC patients. Sorafenib is a multi-target kinase inhibitor that blocks tumor cell proliferation and angiogenesis. Despite sorafenib treatment extending survival, some patients experience side effects, and sorafenib resistance does occur. 3-Hydroxymethyl glutaryl-CoA synthase 2 (HMGCS2) is the rate-limiting enzyme for ketogenesis, which synthesizes the ketone bodies, β-hydroxybutyrate (β-HB) and acetoacetate (AcAc). β-HB is the most abundant ketone body which is present in a 4:1 ratio compared to AcAc. Recently, ketone body treatment was found to have therapeutic effects against many cancers by causing metabolic alternations and cancer cell apoptosis. Our previous publication showed that HMGCS2 downregulation-mediated ketone body reduction promoted HCC clinicopathological progression through regulating c-Myc/cyclin D1 and caspase-dependent signaling. However, whether HMGCS2-regulated ketone body production alters the sensitivity of human HCC to sorafenib treatment remains unclear. In this study, we showed that HMGCS2 downregulation enhanced the proliferative ability and attenuated the cytotoxic effects of sorafenib by activating expressions of phosphorylated (p)-extracellular signal-regulated kinase (ERK), p-P38, and p-AKT. In contrast, HMGCS2 overexpression decreased cell proliferation and enhanced the cytotoxic effects of sorafenib in HCC cells by inhibiting ERK activation. Furthermore, we showed that knockdown HMGCS2 exhibited the potential migratory ability, as well as decreasing zonula occludens protein (ZO)-1 and increasing c-Myc expression in both sorafenib-treated Huh7 and HepG2 cells. Although HMGCS2 overexpression did not alter the migratory effect, expressions of ZO-1, c-Myc, and N-cadherin decreased in sorafenib-treated HMGCS2-overexpressing HCC cells. Finally, we investigated whether ketone treatment influences sorafenib sensitivity. We showed that β-HB pretreatment decreased cell proliferation and enhanced antiproliferative effect of sorafenib in both Huh7 and HepG2 cells. In conclusion, this study defined the impacts of HMGCS2 expression and ketone body treatment on influencing the sorafenib sensitivity of liver cancer cells.
Collapse
Affiliation(s)
- Fat-Moon Suk
- Division of Gastroenterology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chien-Ying Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Wan-Chun Chiu
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Research Center of Geriatric Nutrition, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Department of Nutrition, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chia-Ying Chien
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Tzu-Lang Chen
- Department of Family Medicine, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Yi-Jen Liao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: ; Tel.: +886-2-2736-1661 (ext. 3333)
| |
Collapse
|
35
|
Zhang R, Tomasi D, Shokri-Kojori E, Manza P, Feldman DE, Kroll DS, Biesecker CL, McPherson KL, Schwandt M, Wang GJ, Wiers CE, Volkow ND. Effect of detoxification on N3 sleep correlates with brain functional but not structural changes in alcohol use disorder. Drug Alcohol Depend 2022; 238:109545. [PMID: 35779511 PMCID: PMC9444901 DOI: 10.1016/j.drugalcdep.2022.109545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Sleep disturbances are very common in alcohol use disorder (AUD) and contribute to relapse. Detoxification appears to have limited effects on sleep problems. However, inter-individual differences and related brain mechanisms have not been closely examined. METHODS We examined N3 sleep and the associated brain functional and structural changes in 30 AUD patients (9 Females, mean age: 42 years) undergoing a 3-week inpatient detoxification. Patients' N3 sleep, resting state functional connectivity (RSFC), grey matter volume (GMV) and negative mood were measured on week 1 and week 3. RESULTS AUD patients did not show significant N3 sleep recovery after 3-weeks of detoxification. However, we observed large variability among AUD patients. Inter-individual variations in N3 increases were associated with increases in midline default mode network (DMN) RSFC but not with GMV using a whole-brain approach. Exploratory analyses revealed significant sex by detoxification effects on N3 sleep such that AUD females showed greater N3 increases than AUD males. Further, N3 increases fully mediated the effect of mood improvement on DMN RSFC increases. CONCLUSIONS We show a significant relationship between N3 and DMN functional changes in AUD over time/abstinence. The current findings may have clinical implications for monitoring brain recovery in AUD using daily sleep measures, which might help guide individualized treatments. Future investigations on sex differences with a larger sample and with longitudinal data for a longer period of abstinence are needed.
Collapse
Affiliation(s)
- Rui Zhang
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA.
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Ehsan Shokri-Kojori
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Catherine L Biesecker
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Katherine L McPherson
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Melanie Schwandt
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892-1013, USA; National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892-1013, USA.
| |
Collapse
|
36
|
Edo GI, Nwosu LC. Association of alcohol use and dietary lifestyle of commercial drivers during the COVID-19 pandemic in Nigeria. BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:197. [PMID: 35818411 PMCID: PMC9261201 DOI: 10.1186/s42269-022-00885-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/25/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Alcohol intake, particularly to cope up with stress and depression experienced by commercial drivers during the peak of the COVID-19 pandemic, is alarming as a rise in sales has been reported in certain countries during the quarantine. Alcoholism leads to malnutrition, either because those involved consume an insufficient amount of essential nutrients or because alcohol and its metabolism prevent the body from properly absorbing, digesting, and using those nutrients. This study was carried out to assess the association of alcohol use and dietary lifestyle of commercial motor drivers during the pandemic. RESULTS The anthropometric studies revealed that 69.5% of the respondents fall within the range of 18.5-24.49 indicating that they have normal weights. 63.5% reported daily consumption of alcohol, and 51% claimed that it does not affect their appetite, while 64.5% of the respondent stated that alcohol does not present them with any health problems. There was no significant association between the consumption of alcohol and nutritional status (p > 0.05), but a significant association between dietary lifestyle patterns and the nutritional status of participants was recorded (p < 0.05). CONCLUSION It is, therefore, essential that commercial drivers are given adequate information and guidance on improving their dietary lifestyle.
Collapse
Affiliation(s)
- Great Iruoghene Edo
- Department of Petroleum Chemistry, Faculty of Science, Delta State University of Science & Technology, Ozoro, Nigeria
- Department of Chemistry, Faculty of Arts and Sciences, Cyprus International University, Nicosia, Cyprus
| | - Laurine Chikodiri Nwosu
- Department of Health Care Organizations Management, Faculty of Health Sciences, Cyprus International University, Nicosia, Cyprus
| |
Collapse
|
37
|
Ródenas-González F, Blanco-Gandía MC, Miñarro J, Rodríguez-Arias M. Cognitive profile of male mice exposed to a Ketogenic Diet. Physiol Behav 2022; 254:113883. [PMID: 35716801 DOI: 10.1016/j.physbeh.2022.113883] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 10/18/2022]
Abstract
In recent years, nutritional interventions for different psychiatric diseases have gained increasing attention, such as the ketogenic diet (KD). This has led to positive effects in neurological disorders such as Parkinson's disease, addiction, autism or epilepsy. The neurobiological mechanisms through which these effects are induced and the effects in cognition still warrant investigation, and considering that other high-fat diets (HFD) can lead to cognitive disturbances that may affect the results achieved, the main aim of the present work was to evaluate the effects of a KD to determine whether it can induce such cognitive effects. A total of 30 OF1 male mice were employed to establish the behavioral profile of mice fed a KD by testing anxiety behavior (Elevated Plus Maze), locomotor activity (Open Field), learning (Hebb Williams Maze), and memory (Passive Avoidance Test). The results revealed that the KD did not affect locomotor activity, memory or hippocampal-dependent learning, as similar results were obtained with mice on a standard diet, albeit with increased anxiety behavior. We conclude that a KD is a promising nutritional approach to apply in research studies, given that it does not cause cognitive alterations.
Collapse
Affiliation(s)
- Francisco Ródenas-González
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain
| | - M Carmen Blanco-Gandía
- Departamento de Psicología y Sociología, Facultad de Ciencias Sociales y Humanas, Universidad de Zaragoza, Teruel, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, Valencia, 46010 Spain.
| |
Collapse
|
38
|
Ródenas-González F, Blanco-Gandía MC, Miñarro J, Rodríguez-Arias M. Effects of ketosis on cocaine-induced reinstatement in male mice. Neurosci Lett 2022; 778:136619. [PMID: 35395325 DOI: 10.1016/j.neulet.2022.136619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 10/18/2022]
Abstract
In recent years, the benefits of the ketogenic diet (KD) on different psychiatric disorders have been gaining attention, but the substance abuse field is still unexplored. Some studies have reported that palatable food can modulate the rewarding effects of cocaine, but the negative metabolic consequences rule out the recommendation of using it as a complementary treatment. Thus, the main aim of this study was to evaluate the effects of the KD on cocaine conditioned place preference (CPP) during acquisition, extinction, and reinstatement. 41 OF1 male mice were employed to assess the effects of the KD on a 10 mg/kg cocaine-induced CPP. Animals were divided into three groups: SD, KD, and KD after the Post-Conditioning test. The results revealed that, while access to the KD did not block CPP acquisition, it did significantly reduce the number of sessions required to extinguish the drug-associated memories and it blocked the priming-induced reinstatement.
Collapse
Affiliation(s)
- Francisco Ródenas-González
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - M Carmen Blanco-Gandía
- Departamento de Psicología y Sociología, Facultad de Ciencias Sociales y Humanas, Universidad de Zaragoza, Teruel, Spain
| | - José Miñarro
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Valencia, Spain
| | - Marta Rodríguez-Arias
- Unidad de Investigación Psicobiología de las Drogodependencias, Departamento de Psicobiología, Facultad de Psicología, Universitat de Valencia, Valencia, Spain.
| |
Collapse
|
39
|
Tufvesson-Alm M, Shevchouk OT, Jerlhag E. Insight into the role of the gut-brain axis in alcohol-related responses: Emphasis on GLP-1, amylin, and ghrelin. Front Psychiatry 2022; 13:1092828. [PMID: 36699502 PMCID: PMC9868418 DOI: 10.3389/fpsyt.2022.1092828] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023] Open
Abstract
Alcohol use disorder (AUD) contributes substantially to global morbidity and mortality. Given the heterogenicity of this brain disease, available pharmacological treatments only display efficacy in sub-set of individuals. The need for additional treatment options is thus substantial and is the goal of preclinical studies unraveling neurobiological mechanisms underlying AUD. Although these neurobiological processes are complex and numerous, one system gaining recent attention is the gut-brain axis. Peptides of the gut-brain axis include anorexigenic peptide like glucagon-like peptide-1 (GLP-1) and amylin as well as the orexigenic peptide ghrelin. In animal models, agonists of the GLP-1 or amylin receptor and ghrelin receptor (GHSR) antagonists reduce alcohol drinking, relapse drinking, and alcohol-seeking. Moreover, these three gut-brain peptides modulate alcohol-related responses (behavioral and neurochemical) in rodents, suggesting that the alcohol reduction may involve a suppression of alcohol's rewarding properties. Brain areas participating in the ability of these gut-brain peptides to reduce alcohol-mediated behaviors/neurochemistry involve those important for reward. Human studies support these preclinical studies as polymorphisms of the genes encoding for GLP-1 receptor or the ghrelin pathway are associated with AUD. Moreover, a GLP-1 receptor agonist decreases alcohol drinking in overweight patients with AUD and an inverse GHSR agonist reduces alcohol craving. Although preclinical and clinical studies reveal an interaction between the gut-brain axis and AUD, additional studies should explore this in more detail.
Collapse
Affiliation(s)
- Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Avgerinos KI, Mullins RJ, Egan JM, Kapogiannis D. Ketone Ester Effects on Biomarkers of Brain Metabolism and Cognitive Performance in Cognitively Intact Adults ≥ 55 Years Old. A Study Protocol for a Double-Blinded Randomized Controlled Clinical Trial. J Prev Alzheimers Dis 2022; 9:54-66. [PMID: 35098974 PMCID: PMC9359666 DOI: 10.14283/jpad.2022.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Ketone bodies have been proposed as an "energy rescue" for the Alzheimer's disease (AD) brain, which underutilizes glucose. Prior research has shown that oral ketone monoester (KME) safely induces robust ketosis in humans and has demonstrated cognitive-enhancing and pathology-reducing properties in animal models of AD. However, human evidence that KME may enhance brain ketone metabolism, improve cognitive performance and engage AD pathogenic cascades is scarce. OBJECTIVES To investigate the effects of ketone monoester (KME) on brain metabolism, cognitive performance and AD pathogenic cascades in cognitively normal older adults with metabolic syndrome and therefore at higher risk for AD. DESIGN Double-blinded randomized placebo-controlled clinical trial. SETTING Clinical Unit of the National Institute on Aging, Baltimore, US. PARTICIPANTS Fifty cognitively intact adults ≥ 55 years old, with metabolic syndrome. INTERVENTION Drinks containing 25 g of KME or isocaloric placebo consumed three times daily for 28 days. OUTCOMES Primary: concentration of beta-hydroxybutyrate (BHB) in precuneus measured with Magnetic Resonance Spectroscopy (MRS). Exploratory: plasma and urine BHB, multiple brain and muscle metabolites detected with MRS, cognition assessed with the PACC and NIH toolbox, biomarkers of AD and metabolic mediators in plasma extracellular vesicles, and stool microbiome. DISCUSSION This is the first study to investigate the AD-biomarker and cognitive effects of KME in humans. Ketone monoester is safe, tolerable, induces robust ketosis, and animal studies indicate that it can modify AD pathology. By conducting a study of KME in a population at risk for AD, we hope to bridge the existing gap between pre-clinical evidence and the potential for brain-metabolic, pro-cognitive, and anti-Alzheimer's effects in humans.
Collapse
Affiliation(s)
- K I Avgerinos
- Dimitrios Kapogiannis, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, 8C228, Baltimore, MD 21224, USA; Email address: ; Telephone: +1 410 350 3953
| | | | | | | |
Collapse
|
41
|
Shevchouk OT, Tufvesson-Alm M, Jerlhag E. An Overview of Appetite-Regulatory Peptides in Addiction Processes; From Bench to Bed Side. Front Neurosci 2021; 15:774050. [PMID: 34955726 PMCID: PMC8695496 DOI: 10.3389/fnins.2021.774050] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
There is a substantial need for new pharmacological treatments of addiction, and appetite-regulatory peptides are implied as possible candidates. Appetite regulation is complex and involves anorexigenic hormones such as glucagon-like peptide-1 (GLP-1) and amylin, and orexigenic peptides like ghrelin and all are well-known for their effects on feeding behaviors. This overview will summarize more recent physiological aspects of these peptides, demonstrating that they modulate various aspects of addiction processes. Findings from preclinical, genetic, and experimental clinical studies exploring the association between appetite-regulatory peptides and the acute or chronic effects of addictive drugs will be introduced. Short or long-acting GLP-1 receptor agonists independently attenuate the acute rewarding properties of addictive drugs or reduce the chronic aspects of drugs. Genetic variation of the GLP-1 system is associated with alcohol use disorder. Also, the amylin pathway modulates the acute and chronic behavioral responses to addictive drugs. Ghrelin has been shown to activate reward-related behaviors. Moreover, ghrelin enhances, whereas pharmacological or genetic suppression of the ghrelin receptor attenuates the responses to various addictive drugs. Genetic studies and experimental clinical studies further support the associations between ghrelin and addiction processes. Further studies should explore the mechanisms modulating the ability of appetite-regulatory peptides to reduce addiction, and the effects of combination therapies or different diets on substance use are warranted. In summary, these studies provide evidence that appetite-regulatory peptides modulate reward and addiction processes, and deserve to be investigated as potential treatment target for addiction.
Collapse
Affiliation(s)
- Olesya T Shevchouk
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Maximilian Tufvesson-Alm
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Elisabet Jerlhag
- Department of Pharmacology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
42
|
Kolb H, Kempf K, Röhling M, Lenzen-Schulte M, Schloot NC, Martin S. Ketone bodies: from enemy to friend and guardian angel. BMC Med 2021; 19:313. [PMID: 34879839 PMCID: PMC8656040 DOI: 10.1186/s12916-021-02185-0] [Citation(s) in RCA: 178] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023] Open
Abstract
During starvation, fasting, or a diet containing little digestible carbohydrates, the circulating insulin levels are decreased. This promotes lipolysis, and the breakdown of fat becomes the major source of energy. The hepatic energy metabolism is regulated so that under these circumstances, ketone bodies are generated from β-oxidation of fatty acids and secreted as ancillary fuel, in addition to gluconeogenesis. Increased plasma levels of ketone bodies thus indicate a dietary shortage of carbohydrates. Ketone bodies not only serve as fuel but also promote resistance to oxidative and inflammatory stress, and there is a decrease in anabolic insulin-dependent energy expenditure. It has been suggested that the beneficial non-metabolic actions of ketone bodies on organ functions are mediated by them acting as a ligand to specific cellular targets. We propose here a major role of a different pathway initiated by the induction of oxidative stress in the mitochondria during increased ketolysis. Oxidative stress induced by ketone body metabolism is beneficial in the long term because it initiates an adaptive (hormetic) response characterized by the activation of the master regulators of cell-protective mechanism, nuclear factor erythroid 2-related factor 2 (Nrf2), sirtuins, and AMP-activated kinase. This results in resolving oxidative stress, by the upregulation of anti-oxidative and anti-inflammatory activities, improved mitochondrial function and growth, DNA repair, and autophagy. In the heart, the adaptive response to enhanced ketolysis improves resistance to damage after ischemic insults or to cardiotoxic actions of doxorubicin. Sodium-dependent glucose co-transporter 2 (SGLT2) inhibitors may also exert their cardioprotective action via increasing ketone body levels and ketolysis. We conclude that the increased synthesis and use of ketone bodies as ancillary fuel during periods of deficient food supply and low insulin levels causes oxidative stress in the mitochondria and that the latter initiates a protective (hormetic) response which allows cells to cope with increased oxidative stress and lower energy availability. KEYWORDS: Ketogenic diet, Ketone bodies, Beta hydroxybutyrate, Insulin, Obesity, Type 2 diabetes, Inflammation, Oxidative stress, Cardiovascular disease, SGLT2, Hormesis.
Collapse
Affiliation(s)
- Hubert Kolb
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.,West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| | - Kerstin Kempf
- West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany.
| | - Martin Röhling
- West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| | | | - Nanette C Schloot
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Stephan Martin
- Faculty of Medicine, University of Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.,West-German Centre of Diabetes and Health, Duesseldorf Catholic Hospital Group, Hohensandweg 37, 40591, Duesseldorf, Germany
| |
Collapse
|
43
|
Egervari G, Siciliano CA, Whiteley EL, Ron D. Alcohol and the brain: from genes to circuits. Trends Neurosci 2021; 44:1004-1015. [PMID: 34702580 PMCID: PMC8616825 DOI: 10.1016/j.tins.2021.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/08/2021] [Accepted: 09/30/2021] [Indexed: 01/27/2023]
Abstract
Alcohol use produces wide-ranging and diverse effects on the central nervous system. It influences intracellular signaling mechanisms, leading to changes in gene expression, chromatin remodeling, and translation. As a result of these molecular alterations, alcohol affects the activity of neuronal circuits. Together, these mechanisms produce long-lasting cellular adaptations in the brain that in turn can drive the development and maintenance of alcohol use disorder (AUD). We provide an update on alcohol research, focusing on multiple levels of alcohol-induced adaptations, from intracellular changes to changes in neural circuits. A better understanding of how alcohol affects these diverse and interlinked mechanisms may lead to the identification of novel therapeutic targets and to the development of much-needed novel and efficacious treatment options.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Cell and Developmental Biology, Epigenetics Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Cody A Siciliano
- Department of Pharmacology, Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37203, USA.
| | - Ellanor L Whiteley
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
44
|
Blanco-Gandía MDC, Ródenas-González F, Pascual M, Reguilón MD, Guerri C, Miñarro J, Rodríguez-Arias M. Ketogenic Diet Decreases Alcohol Intake in Adult Male Mice. Nutrients 2021; 13:nu13072167. [PMID: 34202492 PMCID: PMC8308435 DOI: 10.3390/nu13072167] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/19/2021] [Accepted: 06/22/2021] [Indexed: 01/26/2023] Open
Abstract
The classic ketogenic diet is a diet high in fat, low in carbohydrates, and well-adjusted proteins. The reduction in glucose levels induces changes in the body’s metabolism, since the main energy source happens to be ketone bodies. Recent studies have suggested that nutritional interventions may modulate drug addiction. The present work aimed to study the potential effects of a classic ketogenic diet in modulating alcohol consumption and its rewarding effects. Two groups of adult male mice were employed in this study, one exposed to a standard diet (SD, n = 15) and the other to a ketogenic diet (KD, n = 16). When a ketotic state was stable for 7 days, animals were exposed to the oral self-administration paradigm to evaluate the reinforcing and motivating effects of ethanol. Rt-PCR analyses were performed evaluating dopamine, adenosine, CB1, and Oprm gene expression. Our results showed that animals in a ketotic state displayed an overall decrease in ethanol consumption without changes in their motivation to drink. Gene expression analyses point to several alterations in the dopamine, adenosine, and cannabinoid systems. Our results suggest that nutritional interventions may be a useful complementary tool in treating alcohol-use disorders.
Collapse
Affiliation(s)
| | - Francisco Ródenas-González
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (F.R.-G.); (M.P.); (M.D.R.); (J.M.)
| | - María Pascual
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (F.R.-G.); (M.P.); (M.D.R.); (J.M.)
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012 Valencia, Spain;
- Department of Physiology, School of Medicine, Universitat de Valencia, Avda. Blasco Ibáñez, 15, 46010 Valencia, Spain
| | - Marina Daiana Reguilón
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (F.R.-G.); (M.P.); (M.D.R.); (J.M.)
| | - Consuelo Guerri
- Department of Molecular and Cellular Pathology of Alcohol, Principe Felipe Research Center, C/Eduardo Primo Yúfera 3, 46012 Valencia, Spain;
| | - José Miñarro
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (F.R.-G.); (M.P.); (M.D.R.); (J.M.)
| | - Marta Rodríguez-Arias
- Unit of Research Psychobiology of Drug Dependence, Department of Psychobiology, Facultad de Psicología, Universitat de Valencia, Avda. Blasco Ibáñez, 21, 46010 Valencia, Spain; (F.R.-G.); (M.P.); (M.D.R.); (J.M.)
- Correspondence: ; Tel.: +34-963864637
| |
Collapse
|
45
|
Mahajan VR, Elvig SK, Vendruscolo LF, Koob GF, Darcey VL, King MT, Kranzler HR, Volkow ND, Wiers CE. Nutritional Ketosis as a Potential Treatment for Alcohol Use Disorder. Front Psychiatry 2021; 12:781668. [PMID: 34916977 PMCID: PMC8670944 DOI: 10.3389/fpsyt.2021.781668] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/08/2021] [Indexed: 12/28/2022] Open
Abstract
Alcohol use disorder (AUD) is a chronic, relapsing brain disorder, characterized by compulsive alcohol seeking and disrupted brain function. In individuals with AUD, abstinence from alcohol often precipitates withdrawal symptoms than can be life threatening. Here, we review evidence for nutritional ketosis as a potential means to reduce withdrawal and alcohol craving. We also review the underlying mechanisms of action of ketosis. Several findings suggest that during alcohol intoxication there is a shift from glucose to acetate metabolism that is enhanced in individuals with AUD. During withdrawal, there is a decline in acetate levels that can result in an energy deficit and could contribute to neurotoxicity. A ketogenic diet or ingestion of a ketone ester elevates ketone bodies (acetoacetate, β-hydroxybutyrate and acetone) in plasma and brain, resulting in nutritional ketosis. These effects have been shown to reduce alcohol withdrawal symptoms, alcohol craving, and alcohol consumption in both preclinical and clinical studies. Thus, nutritional ketosis may represent a unique treatment option for AUD: namely, a nutritional intervention that could be used alone or to augment the effects of medications.
Collapse
Affiliation(s)
- Vikrant R Mahajan
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States
| | - Sophie K Elvig
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Leandro F Vendruscolo
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - George F Koob
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, Baltimore, MD, United States
| | - Valerie L Darcey
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - M Todd King
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Henry R Kranzler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Rockville, MD, United States
| | - Corinde E Wiers
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
46
|
Alcohol. Alcohol 2021. [DOI: 10.1016/b978-0-12-816793-9.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|