1
|
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
|
2
|
Cao J, Ball IK, Summerell E, Humburg P, Denson T, Rae CD. Effect of Ethanol on Brain Electrical Tissue Conductivity in Social Drinkers. J Magn Reson Imaging 2025; 61:1181-1187. [PMID: 39105662 PMCID: PMC11803702 DOI: 10.1002/jmri.29548] [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: 04/16/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND How the biophysics of electrical conductivity measures relate to brain activity is poorly understood. The sedative, ethanol, reduces metabolic activity but its impact on brain electrical conductivity is unknown. PURPOSE To investigate whether ethanol reduces brain electrical tissue conductivity. STUDY TYPE Prospective. SUBJECTS Fifty-two healthy volunteers (aged 18-37 years, 22 females, 30 males). FIELD STRENGTH/SEQUENCE 3 T, T1-weighted, multi-shot, turbo-field echo (TFE); 3D balanced fast-field echo (bFFE). ASSESSMENT Brain gray and white matter tissue conductivity measured with phase-based magnetic resonance electrical properties tomography (MREPT) compared before and 20 minutes after ethanol consumption (0.7 g/kg body weight). Differential conductivity whole brain maps were generated for three subgroups: those with strong ( ∆ σ max > 0.1 S/m; N = 33), weak (0.02 S/m ≤ ∆ σ max ≤ 0.1 S/m; N = 9) conductivity decrease, and no significant response ( ∆ σ max < 0.02 S/m, N = 10). Maps were compared in the strong response group where breath alcohol rose between scans, vs. those where it fell. STATISTICAL TESTS Average breath alcohol levels were compared to the differential conductivity maps using linear regression. T-maps were generated (threshold P < 0.05 and P < 0.001; minimum cluster 48 mm3). Differential conductivity maps were compared with ANOVA. RESULTS Whole-group analysis showed decreased conductivity that did not survive statistical thresholding. Strong responders (N = 33) showed a consistent pattern of significantly decreased conductivity ( ∆ σ max > 0.1 S/m) in frontal/occipital and cerebellar white matter. The weak response group (N = 9) showed a similar pattern of conductivity decrease (0.02 S/m ≤ ∆ σ max ≤ 0.1 S/m). There was no significant relationship with breath alcohol levels, alcohol use, age, ethnicity, or sex. The strong responders' regional response was different between ascending (N = 12) or descending (N = 20) alcohol during the scan. DATA CONCLUSION Ethanol reduces brain tissue conductivity in a participant-dependent and spatially dependent fashion. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jun Cao
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
| | - Iain K. Ball
- Philips Australia & New ZealandNorth RydeNew South WalesAustralia
| | - Elizabeth Summerell
- School of Psychology, The University of New South WalesSydneyNew South WalesAustralia
| | - Peter Humburg
- Mark Wainwright Analytical Centre, Stats Central, The University of New South WalesSydneyNew South WalesAustralia
| | - Tom Denson
- School of Psychology, The University of New South WalesSydneyNew South WalesAustralia
| | - Caroline D. Rae
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- School of Psychology, The University of New South WalesSydneyNew South WalesAustralia
| |
Collapse
|
3
|
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
|
4
|
Plantone D, Stufano A, Righi D, Locci S, Iavicoli I, Lovreglio P, De Stefano N. Neurofilament light chain and glial fibrillary acid protein levels are elevated in post-mild COVID-19 or asymptomatic SARS-CoV-2 cases. Sci Rep 2024; 14:6429. [PMID: 38499607 PMCID: PMC10948776 DOI: 10.1038/s41598-024-57093-z] [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: 10/17/2023] [Accepted: 03/14/2024] [Indexed: 03/20/2024] Open
Abstract
Given the huge impact of the COVID-19 pandemic, it appears of paramount importance to assess the cognitive effects on the population returning to work after COVID-19 resolution. Serum levels of neurofilament light chain (sNfL) and glial fibrillary acidic protein (sGFAP) represent promising biomarkers of neuro-axonal damage and astrocytic activation. In this cohort study, we explored the association between sNfL and sGFAP concentrations and cognitive performance in a group of 147 adult workers with a previous asymptomatic SARS-CoV-2 infection or mild COVID-19, one week and, in 49 of them, ten months after SARS-Cov2 negativization and compared them to a group of 82 age and BMI-matched healthy controls (HCs). sNfL and sGFAP concentrations were assessed using SimoaTM assay Neurology 2-Plex B Kit. COVID-19 patients were interviewed one-on-one by trained physicians and had to complete a list of questionnaires, including the Cognitive Failure Questionnaire (CFQ). At the first assessment (T0), sNfL and sGFAP levels were significantly higher in COVID-19 patients than in HCs (p < 0.001 for both). The eleven COVID-19 patients with cognitive impairment had significantly higher levels of sNfL and sGFAP than the others (p = 0.005 for both). At the subsequent follow-up (T1), sNfL and sGFAP levels showed a significant decrease (median sNfL 18.3 pg/mL; median sGFAP 77.2 pg/mL), although they were still higher than HCs (median sNfL 7.2 pg/mL, median sGFAP 63.5 pg/mL). Our results suggest an ongoing damage involving neurons and astrocytes after SARS-Cov2 negativization, which reduce after ten months even if still evident compared to HCs.
Collapse
Affiliation(s)
- Domenico Plantone
- Department of Medicine, Surgery, Neuroscience University of Siena, Siena, Italy.
| | - Angela Stufano
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Delia Righi
- Department of Medicine, Surgery, Neuroscience University of Siena, Siena, Italy
| | - Sara Locci
- Department of Medicine, Surgery, Neuroscience University of Siena, Siena, Italy
| | - Ivo Iavicoli
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Piero Lovreglio
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery, Neuroscience University of Siena, Siena, Italy
| |
Collapse
|
5
|
Popova D, Sun J, Chow HM, Hart RP. A critical review of ethanol effects on neuronal firing: A metabolic perspective. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:450-458. [PMID: 38217065 PMCID: PMC10966925 DOI: 10.1111/acer.15266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Ethanol metabolism is relatively understudied in neurons, even though changes in neuronal metabolism are known to affect their activity. Recent work demonstrates that ethanol is preferentially metabolized over glucose as a source of carbon and energy, and it reprograms neurons to a state of reduced energy potential and diminished capacity to utilize glucose once ethanol is exhausted. Ethanol intake has been associated with changes in neuronal firing and specific brain activity (EEG) patterns have been linked with risk for alcohol use disorder (AUD). Furthermore, a haplotype of the inwardly rectifying potassium channel subunit, GIRK2, which plays a critical role in regulating excitability of neurons, has been linked with AUD and shown to be directly regulated by ethanol. At the same time, overexpression of GIRK2 prevents ethanol-induced metabolic changes. Based on the available evidence, we conclude that the mechanisms underlying the effects of ethanol on neuronal metabolism are a novel target for developing therapies for AUD.
Collapse
Affiliation(s)
- Dina Popova
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
- Present address: Neuroscience Institute, NYU Langone Grossman School of Medicine, New York, NY USA
| | - Jacquelyne Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ronald P. Hart
- Department of Cell Biology & Neuroscience, Rutgers University, Piscataway NJ USA
| |
Collapse
|
6
|
Boissoneault J, Stennett-Blackmon B, Gilmour C, Blaes S. Neural and Psychosocial Mechanisms Underlying Alcohol Use and Pain Interactions: Overview of Current Evidence and Future Directions. CURRENT ADDICTION REPORTS 2023; 10:677-689. [PMID: 38645279 PMCID: PMC11031255 DOI: 10.1007/s40429-023-00518-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 04/23/2024]
Abstract
Purpose of Review A growing body of research indicates bidirectional associations between alcohol use and pain. In this review, we highlight common neural and psychosocial mechanisms underlying pain and alcohol use and identify current gaps in the literature regarding alcohol/pain interactions. We also suggest future directions for the field moving forward, including more nuanced conceptualization of alcohol's negative reinforcing effects in the context of pain, broader use of clinically-relevant experimental pain induction modalities, and characterization of age, biological sex, gender, race, and ethnicity as moderators of pain/alcohol interactions. Recent Findings Acute alcohol intake has analgesic and negative-reinforcing effects in the context of pain, and chronic heavy alcohol use appears to increase risk for development of chronic pain. At the same time, pain, both acute and chronic, acts as a proximal antecedent for alcohol use and is associated with relapse risk for individuals in recovery from alcohol use disorder. Summary Although the links between alcohol use and pain are increasingly appreciated, significant gaps in understanding remain and systematic study of alcohol/pain interactions at all levels, including basic, preclinical, translational, and interventional, is needed.
Collapse
Affiliation(s)
- Jeff Boissoneault
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, USA
- Department of Anesthesiology, University of Minnesota, Minneapolis, MN, USA
| | - Bethany Stennett-Blackmon
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, USA
| | - Christina Gilmour
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, USA
| | - Shelby Blaes
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, USA
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Huang YH, Lee MT, Hsueh HY, Knutson DE, Cook J, Mihovilovic MD, Sieghart W, Chiou LC. Cerebellar α6GABA A Receptors as a Therapeutic Target for Essential Tremor: Proof-of-Concept Study with Ethanol and Pyrazoloquinolinones. Neurotherapeutics 2023; 20:399-418. [PMID: 36696034 PMCID: PMC10121996 DOI: 10.1007/s13311-023-01342-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2022] [Indexed: 01/26/2023] Open
Abstract
Ethanol has been shown to suppress essential tremor (ET) in patients at low-to-moderate doses, but its mechanism(s) of action remain unknown. One of the ET hypotheses attributes the ET tremorgenesis to the over-activated firing of inferior olivary neurons, causing synchronic rhythmic firings of cerebellar Purkinje cells. Purkinje cells, however, also receive excitatory inputs from granule cells where the α6 subunit-containing GABAA receptors (α6GABAARs) are abundantly expressed. Since ethanol is a positive allosteric modulator (PAM) of α6GABAARs, such action may mediate its anti-tremor effect. Employing the harmaline-induced ET model in male ICR mice, we evaluated the possible anti-tremor effects of ethanol and α6GABAAR-selective pyrazoloquinolinone PAMs. The burrowing activity, an indicator of well-being in rodents, was measured concurrently. Ethanol significantly and dose-dependently attenuated action tremor at non-sedative doses (0.4-2.4 g/kg, i.p.). Propranolol and α6GABAAR-selective pyrazoloquinolinones also significantly suppressed tremor activity. Neither ethanol nor propranolol, but only pyrazoloquinolinones, restored burrowing activity in harmaline-treated mice. Importantly, intra-cerebellar micro-injection of furosemide (an α6GABAAR antagonist) had a trend of blocking the effect of pyrazoloquinolinone Compound 6 or ethanol on harmaline-induced tremor. In addition, the anti-tremor effects of Compound 6 and ethanol were synergistic. These results suggest that low doses of ethanol and α6GABAAR-selective PAMs can attenuate action tremor, at least partially by modulating cerebellar α6GABAARs. Thus, α6GABAARs are potential therapeutic targets for ET, and α6GABAAR-selective PAMs may be a potential mono- or add-on therapy.
Collapse
Affiliation(s)
- Ya-Hsien Huang
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan
| | - Ming Tatt Lee
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan
- Faculty of Pharmaceutical Sciences, UCSI University, Kuala Lumpur, 56000, Malaysia
| | - Han-Yun Hsueh
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan
| | - Daniel E Knutson
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - James Cook
- Department of Chemistry and Biochemistry, Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | | | - Werner Sieghart
- Center for Brain Research, Department of Molecular Neurosciences, Medical University Vienna, Vienna, 1090, Austria
| | - Lih-Chu Chiou
- Department and Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, No. 1, Jen-Ai Rd., Section 1, Taipei, 10051, Taiwan.
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
8
|
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
|
9
|
van den Oord EJCG, Xie LY, Zhao M, Aberg KA, Clark SL. A single-nucleus transcriptomics study of alcohol use disorder in the nucleus accumbens. Addict Biol 2023; 28:e13250. [PMID: 36577731 DOI: 10.1111/adb.13250] [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: 04/15/2022] [Revised: 08/29/2022] [Accepted: 10/13/2022] [Indexed: 11/14/2022]
Abstract
Gene expression studies offer promising opportunities to better understand the processes underlying alcohol use disorder (AUD). As cell types differ in their function, gene expression profiles will typically vary across cell types. When studying bulk tissue, failure to account for this cellular diversity has a detrimental impact on the ability to detect disease associations. We therefore assayed the transcriptomes of 32,531 individual nuclei extracted from the nucleus accumbens (NAc) of nine donors with AUD and nine controls (72% male). Our study identified 17 clearly delineated cell types. We detected 26 transcriptome-wide significant differentially expressed genes (DEGs) that mainly involved medium spiny neurons with both D1-type and D2-type dopamine receptors, microglia (MGL) and oligodendrocytes. A higher than expected number of DEGs replicated in an existing single nucleus gene expression study of alcohol dependence in the prefrontal cortex (enrichment ratio 1.91, p value 0.019) with two genes remaining significant after a Bonferroni correction. Our most compelling result involved CD53 in MGL that replicated in the same cell type in the prefrontal cortex and was previously implicated in studies of DNA methylation, bulk gene expression and genetic variants. Several DEGs were previously reported to be associated with AUD (e.g., PER1 and MGAT5). The DEGs for MSN.3 seemed involved in neurodegeneration, disruption of circadian rhythms, alterations in glucose metabolism and changes in synaptic plasticity. For MGL, the DEGs implicated neuroinflammation and immune-related processes and for OLI, disruptions in myelination. This identification of the specific cell-types from which the association signals originate will be key for designing proper follow-up experiments and, eventually, novel clinical interventions.
Collapse
Affiliation(s)
- Edwin J C G van den Oord
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Lin Y Xie
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Min Zhao
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Karolina A Aberg
- Center for Biomarker Research and Precision Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Shaunna L Clark
- Department of Psychiatry & Behavioral Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
10
|
Hamel R, Demers O, Boileau C, Roy ML, Théoret H, Bernier PM, Lepage JF. The neurobiological markers of acute alcohol's subjective effects in humans. Neuropsychopharmacology 2022; 47:2101-2110. [PMID: 35701548 PMCID: PMC9556716 DOI: 10.1038/s41386-022-01354-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 12/21/2022]
Abstract
The ingestion of alcohol yields acute biphasic subjective effects: stimulation before sedation. Despite their predictive relevance to the development of alcohol use disorders (AUD), the neurobiological markers accounting for the biphasic effects of alcohol remain poorly understood in humans. Informed by converging lines of evidence, this study tested the hypothesis that alcohol ingestion acutely increases gamma-aminobutyric acid (GABA)-mediated inhibition, which would positively and negatively predict the feeling of stimulation and sedation, respectively. To do so, healthy participants (n = 20) ingested a single dose of 94% ABV alcohol (males: 1.0 ml/kg; females: 0.85 ml/kg) in a randomized placebo-controlled cross-over design. The alcohol's biphasic effects were assessed with the Brief-Biphasic Alcohol Effects Scale, and non-invasive neurobiological markers were measured with transcranial magnetic stimulation, before and every 30 min (up to 120 min) after the complete ingestion of the beverage. Results showed that acute alcohol ingestion selectively increased the duration of the cortical silent period (CSP) as compared to placebo, suggesting that alcohol increases non-specific GABAergic inhibition. Importantly, CSP duration positively and negatively predicted increases in the feeling of stimulation and sedation, respectively, suggesting that stimulation emerges as GABAergic inhibition increases and that sedation emerges as GABAergic inhibition returns to baseline values. Overall, these results suggest that modulations of GABAergic inhibition are central to the acute biphasic subjective effects of alcohol, providing a potential preventive target to curb the progression of at-risk individuals to AUD.
Collapse
Affiliation(s)
- Raphael Hamel
- Département de kinanthropologie, Faculté des sciences de l'activité physique, Université de Sherbrooke, Sherbrooke, QC, Canada
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Demers
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Camille Boileau
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Marie-Laurence Roy
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Théoret
- Département de psychologie, Faculté des arts et sciences, Université de Montréal, Montreal, QC, Canada
| | - Pierre-Michel Bernier
- Département de kinanthropologie, Faculté des sciences de l'activité physique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Francois Lepage
- Département de pédiatrie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
- Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
11
|
Rapp C, Hamilton J, Richer K, Sajjad M, Yao R, Thanos PK. Alcohol binge drinking decreases brain glucose metabolism and functional connectivity in adolescent rats. Metab Brain Dis 2022; 37:1901-1908. [PMID: 35567647 DOI: 10.1007/s11011-022-00977-8] [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: 09/29/2021] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
Abstract
Alcohol misuse represents a serious health concern, especially during adolescence, with approximately 18% of high school students engaging in binge drinking. Despite widespread misuse of alcohol, its effects on how the brain functions is not fully understood. This study utilized a binge drinking model in adolescent rats to examine effects on brain function as measured by brain glucose metabolism (BGluM). Following an injection of [18 FDG] fluro-2-deoxy-D-glucose, rats had voluntary access to either water or various concentrations of ethanol to obtain the following targeted doses: water (no ethanol), low dose ethanol (0.29 ± 0.03 g/kg), moderate dose ethanol (0.98 ± 0.05), and high dose ethanol (2.19 ± 0.23 g/kg). Rats were subsequently scanned using positron emission tomography. All three doses of ethanol were found to decrease BGluM in the restrosplenial cortex, visual cortex, jaw region of the somatosensory cortex, and cerebellum. For both the LD and MD ethanol dose, decreased BGluM was seen in the superior colliculi. The MD ethanol dose also decreased BGluM in the subiculum, frontal association area, as well as the primary motor cortex. Lastly, the HD ethanol dose decreased BGluM in the hippocampus, thalamus, raphe nucleus, inferior colliculus, and the primary motor cortex. Similar decreases in the hippocampus were also seen in the LD group. Taken together, these results highlight the negative consequences of acute binge drinking on BGluM in many regions of the brain involved in sensory, motor, and cognitive processes. Future studies are needed to assess the long-term effects of alcohol binge drinking on brain function as well as its cessation.
Collapse
Affiliation(s)
- Cecilia Rapp
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical and Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, United States
- Department of Biomedical Engineering, State University at New York at Buffalo, Buffalo, NY, United States
| | - John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical and Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, United States
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York, United States
| | - Kaleigh Richer
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical and Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, United States
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York, United States
| | - Munawwar Sajjad
- Department of Nuclear Medicine, University at Buffalo, Buffalo, United States
| | - Rutao Yao
- Department of Nuclear Medicine, University at Buffalo, Buffalo, United States
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical and Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY, United States.
- Department of Psychology, State University of New York at Buffalo, Buffalo, New York, United States.
| |
Collapse
|
12
|
Vitus D, Williams MK, Rizk M, Neubert JK, Robinson M, Boissoneault J. Analgesic effects of alcohol in adults with chronic jaw pain. Alcohol Clin Exp Res 2022; 46:1515-1524. [PMID: 35989585 PMCID: PMC9427671 DOI: 10.1111/acer.14883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Although recent literature provides promising support for the analgesic properties of alcohol, potential differences in alcohol analgesia as a function of chronic pain status are not well understood. Thus, this study examined chronic pain status as a potential moderator of alcohol analgesia and distinguished between multiple aspects of pain experience and sensitivity: pain threshold, pain intensity, pain unpleasantness, and perceived relief. METHODS Social drinkers with (N = 19) and without (N = 29) chronic jaw pain completed two testing sessions in a counterbalanced order: alcohol (target BrAC = 0.08 g/dl) and placebo. In each, pressure algometry was performed at the insertion of the masseter. Alcohol analgesia was assessed by examining the main and interactive effects of beverage condition, pressure level (4, 5, or 6 pound-feet [lbf]), and chronic jaw pain status (chronic pain vs. pain-free control) on quantitative sensory testing measures and pain relief ratings following noxious stimuli. RESULTS Analyses indicated significant increases in pain threshold and pain relief and reductions in pain unpleasantness and pain intensity, under the alcohol condition. Chronic pain participants demonstrated lower pain thresholds and greater pain intensity and pain unpleasantness ratings than controls. There were no interactive effects of alcohol and pain conditions on any pain measure. CONCLUSIONS Findings provide experimental evidence of alcohol's analgesic and pain-relieving effects and suggest that these effects do not significantly differ by chronic pain status. Individuals, who self-medicate pain via alcohol consumption, irrespective of pain status, may be at increased risk to engage in hazardous drinking patterns and thus experience adverse alcohol-related consequences.
Collapse
Affiliation(s)
- Darya Vitus
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32610
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
| | - Michelle K. Williams
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32610
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
| | - Mehdi Rizk
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32610
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
| | - John K. Neubert
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
- Department of Orthodontics, University of Florida, Gainesville, FL, 32610
| | - Michael Robinson
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32610
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
| | - Jeff Boissoneault
- Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, 32610
- Center for Pain Research and Behavioral Health, University of Florida, Gainesville, FL, 32610
| |
Collapse
|
13
|
Zhong L, Chen W, Wang T, Zeng Q, Lai L, Lai J, Lin J, Tang S. Alcohol and Health Outcomes: An Umbrella Review of Meta-Analyses Base on Prospective Cohort Studies. Front Public Health 2022; 10:859947. [PMID: 35602135 PMCID: PMC9115901 DOI: 10.3389/fpubh.2022.859947] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 04/06/2022] [Indexed: 12/18/2022] Open
Abstract
An umbrella review of meta-analyses was performed to summarize the evidence of associations between alcohol consumption and health outcomes and to assess its credibility. Meta-analyses of prospective cohort studies reporting the associations of alcohol consumption with health outcomes were identified. We recalculated the random-effects summary effect size and 95% confidence interval, heterogeneity, and small-study effect for each meta-analysis and graded the evidence. Fifty-nine publications reporting 224 meta-analyses of prospective cohort studies with 140 unique health outcomes were included, in which there were 49 beneficial associations and 25 harmful associations with nominally statistically significant summary results. But quality of evidence was rated high only for seven beneficial associations (renal cell carcinoma risk, dementia risk, colorectal cancer mortality, and all-cause mortality in patients with hypertension for low alcohol consumption; renal cell carcinoma risk, cardiovascular disease (CVD) risk in patients with hypertension and all-cause mortality in patients with hypertension for moderate consumption) and four harmful associations (cutaneous basal cell carcinoma risk for low alcohol consumption; cutaneous basal cell carcinoma risk and cutaneous squamous cell carcinoma risk for moderate alcohol consumption; hemorrhagic stroke risk for high alcohol consumption). In this umbrella review, only 11 health outcomes (5 in low alcohol consumption, 5 in moderate alcohol consumption and 1 in high alcohol consumption) with statistically significant showed high quality of epidemiologic evidence. More robust and larger prospective studies are needed to verify our results.
Collapse
Affiliation(s)
- Lixian Zhong
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Weiwei Chen
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Gastroenterology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Tonghua Wang
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Qiuting Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Leizhen Lai
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Junlong Lai
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Junqin Lin
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
14
|
Randall PA, Lovelock DF, VanVoorhies K, Agan VE, Kash TL, Besheer J. Low-dose alcohol: Interoceptive and molecular effects and the role of dentate gyrus in rats. Addict Biol 2021; 26:e12965. [PMID: 33015936 DOI: 10.1111/adb.12965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 01/24/2023]
Abstract
Alcohol abuse and dependence are world-wide health problems. Most research on alcohol use focuses on the consequences of moderate to high levels of alcohol. However, even at low concentrations, alcohol is capable of producing effects in the brain that can ultimately affect behavior. The current studies seek to understand the effects of low-dose alcohol (blood alcohol levels of ≤10mM). To do so, these experiments utilize a combination of behavioral and molecular techniques to (1) assess the ability of the interoceptive effects of a low dose of alcohol to gain control over goal-tracking behavior in a Pavlovian discrimination task, (2) determine brain regional differences in cellular activity via expression of immediate early genes (IEGs), and (3) assess the role of the dentate gyrus in modulating sensitivity to the interoceptive effects of a low dose of alcohol. Here, we show that intragastric administration of a dose of 0.8 g/kg alcohol produces blood alcohol levels ≤10mM in both male and female Long-Evans rats and can readily be trained as a Pavlovian interoceptive drug cue. In rats trained on this procedure, this dose of alcohol also modulates expression of the IEGs c-Fos and Arc in brain regions known to modulate expression of alcohol interoceptive effects. Finally, pharmacological inactivation of the dentate gyrus with GABA agonists baclofen and muscimol disrupted the ability of a low dose of alcohol to serve as an interoceptive cue. Together, these findings demonstrate behavioral and molecular consequences of low-dose alcohol.
Collapse
Affiliation(s)
- Patrick A. Randall
- Department of Anesthesiology and Perioperative Medicine Penn State College of Medicine Hershey Pennsylvania USA
- Department of Pharmacology Penn State College of Medicine Hershey Pennsylvania USA
| | - Dennis F. Lovelock
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Kalynn VanVoorhies
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Verda E. Agan
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Pharmacology University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - Joyce Besheer
- Bowles Center for Alcohol Studies University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
- Department of Psychiatry University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| |
Collapse
|
15
|
Tanabe J, Neff S, Sutton B, Ellis S, Patten L, Brown MS, Hoffman PL, Tabakoff B, Burnham EL. Effects of acetate on cerebral blood flow, systemic inflammation, and behavior in alcohol use disorder. Alcohol Clin Exp Res 2021; 45:922-933. [PMID: 33682145 PMCID: PMC8496991 DOI: 10.1111/acer.14588] [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: 11/10/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol use disorders (AUDs) are associated with altered regulation of physiological processes in the brain. Acetate, a metabolite of ethanol, has been implicated in several processes that are disrupted in AUDs including transcriptional regulation, metabolism, inflammation, and neurotransmission. To further understand the effects of acetate on brain function in AUDs, we investigated the effects of acetate on cerebral blood flow (CBF), systemic inflammatory cytokines, and behavior in AUD. METHODS Sixteen participants with AUD were recruited from a nonmedical, clinically managed detoxification center. Each participant received acetate and placebo in a randomly assigned order of infusion and underwent 3T MR scanning using quantitative pseudo-continuous arterial spin labeling. Participants and the study team were blinded to the infusion. CBF values (ml/100 g/min) extracted from thalamus were compared between placebo and acetate using a mixed effect linear regression model accounting for infusion order. Voxel-wise CBF comparisons were set at threshold of p < 0.05 cluster-corrected for multiple comparisons, voxel-level p < 0.0001. Plasma cytokine levels and behavior were also assessed between infusions. RESULTS Fifteen men and 1 woman were enrolled with Alcohol Use Disorders Identification Test (AUDIT) scores between 13 and 38 with a mean of 28.3 ± 9.1. Compared to placebo, acetate administration increased CBF in the thalamus bilaterally (Left: 51.2 vs. 68.8, p < 0.001; Right: 53.7 vs. 69.6, p = 0.001), as well as the cerebellum, brainstem, and cortex. Older age and higher AUDIT scores were associated with increases in acetate-induced thalamic blood flow. Cytokine levels and behavioral measures did not differ between placebo and acetate infusions. CONCLUSIONS This pilot study in AUD suggests that during the first week of abstinence from alcohol, the brain's response to acetate differs by brain region and this response may be associated with the severity of alcohol dependence.
Collapse
Affiliation(s)
- Jody Tanabe
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Sarah Neff
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Brianne Sutton
- Department of Psychiatry, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Sam Ellis
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Luke Patten
- Department of Biostatistics and Informatics, School of Public Health; University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Mark S. Brown
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Paula L. Hoffman
- Department of Pharmacology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| | - Ellen L. Burnham
- Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045
| |
Collapse
|
16
|
Effects of ketogenic diet and ketone monoester supplement on acute alcohol withdrawal symptoms in male mice. Psychopharmacology (Berl) 2021; 238:833-844. [PMID: 33410985 PMCID: PMC7914216 DOI: 10.1007/s00213-020-05735-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022]
Abstract
RATIONALE After alcohol ingestion, the brain partly switches from consumption of glucose to consumption of the alcohol metabolite acetate. In heavy drinkers, the switch persists after abrupt abstinence, leading to the hypothesis that the resting brain may be "starved" when acetate levels suddenly drop during abstinence, despite normal blood glucose, contributing to withdrawal symptoms. We hypothesized that ketone bodies, like acetate, could act as alternative fuels in the brain and alleviate withdrawal symptoms. OBJECTIVES We previously reported that a ketogenic diet during alcohol exposure reduced acute withdrawal symptoms in rats. Here, our goals were to test whether (1) we could reproduce our findings, in mice and with longer alcohol exposure; (2) ketone bodies alone are sufficient to reduce withdrawal symptoms (clarifying mechanism); (3) introduction of ketogenic diets at abstinence (a clinically more practical implementation) would also be effective. METHODS Male C57BL/6NTac mice had intermittent alcohol exposure for 3 weeks using liquid diet. Somatic alcohol withdrawal symptoms were measured as handling-induced convulsions; anxiety-like behavior was measured using the light-dark transition test. We tested a ketogenic diet, and a ketone monoester supplement with a regular carbohydrate-containing diet. RESULTS The regular diet with ketone monoester was sufficient to reduce handling-induced convulsions and anxiety-like behaviors in early withdrawal. Only the ketone monoester reduced handling-induced convulsions when given during abstinence, consistent with faster elevation of blood ketones, relative to ketogenic diet. CONCLUSIONS These findings support the potential utility of therapeutic ketosis as an adjunctive treatment in early detoxification in alcohol-dependent patients seeking to become abstinent. TRIAL REGISTRATION clinicaltrials.gov NCT03878225, NCT03255031.
Collapse
|
17
|
Bordia T, Zahr NM. The Inferior Colliculus in Alcoholism and Beyond. Front Syst Neurosci 2020; 14:606345. [PMID: 33362482 PMCID: PMC7759542 DOI: 10.3389/fnsys.2020.606345] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/02/2020] [Indexed: 12/28/2022] Open
Abstract
Post-mortem neuropathological and in vivo neuroimaging methods have demonstrated the vulnerability of the inferior colliculus to the sequelae of thiamine deficiency as occurs in Wernicke-Korsakoff Syndrome (WKS). A rich literature in animal models ranging from mice to monkeys-including our neuroimaging studies in rats-has shown involvement of the inferior colliculi in the neural response to thiamine depletion, frequently accomplished with pyrithiamine, an inhibitor of thiamine metabolism. In uncomplicated alcoholism (i.e., absent diagnosable neurological concomitants), the literature citing involvement of the inferior colliculus is scarce, has nearly all been accomplished in preclinical models, and is predominately discussed in the context of ethanol withdrawal. Our recent work using novel, voxel-based analysis of structural Magnetic Resonance Imaging (MRI) has demonstrated significant, persistent shrinkage of the inferior colliculus using acute and chronic ethanol exposure paradigms in two strains of rats. We speculate that these consistent findings should be considered from the perspective of the inferior colliculi having a relatively high CNS metabolic rate. As such, they are especially vulnerable to hypoxic injury and may be provide a common anatomical link among a variety of disparate insults. An argument will be made that the inferior colliculi have functions, possibly related to auditory gating, necessary for awareness of the external environment. Multimodal imaging including diffusion methods to provide more accurate in vivo visualization and quantification of the inferior colliculi may clarify the roles of brain stem nuclei such as the inferior colliculi in alcoholism and other neuropathologies marked by altered metabolism.
Collapse
Affiliation(s)
- Tanuja Bordia
- Neuroscience Program, SRI International, Menlo Park, CA, United States
| | - Natalie M. Zahr
- Neuroscience Program, SRI International, Menlo Park, CA, United States
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
18
|
Abstract
Patients with essential tremor, vocal tremor, torticollis, myoclonus-dystonia and posthypoxic myoclonus often benefit in a surprisingly rapid and robust manner from ingestion of a modest amount of alcohol (ethanol). Despite considerable investigation, the mechanism of ethanol’s ability to produce this effect remains a mystery. In this paper, we review the pharmacology of ethanol and its analogue GHB (or sodium oxybate), summarize the published literature of alcohol-responsive hyperkinetic movement disorders, and demonstrate videos of patients we have treated over the last fifteen years with either an ethanol challenge or with chronic sodium oxybate therapy. We then propose a novel explanation for this phenomenon—namely, that ingestion of modest doses of ethanol (or sodium oxybate) normalizes the aberrant motor networks underling these disorders. We propose that alcohol and its analogues improve clinical symptoms and their physiologic correlate by restoring the normal firing pattern of the major outflow pathways of the cerebellum (the Purkinje cells and deep cerebellar nuclei), We present evidence to support this hypothesis in animal models and in affected patients, and suggest future investigations to test this model.
Collapse
|
19
|
Wilson DF, Matschinsky FM. Ethanol metabolism: The good, the bad, and the ugly. Med Hypotheses 2020; 140:109638. [PMID: 32113062 DOI: 10.1016/j.mehy.2020.109638] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022]
Abstract
Throughout the world, ethanol is both an important commercial commodity and a source of major medical and social problems. Ethanol readily passes through biological membranes and distributes throughout the body. It is oxidized, first to acetaldehyde and then to acetate, and finally by the citric acid cycle in virtually all tissues. The oxidation of ethanol is irreversible and unregulated, making the rate dependent only on local concentration and enzyme activity. This unregulated input of reducing equivalents increases reduction of both cytoplasmic and intramitochondrial NAD and, through the latter, cellular energy state {[ATP]/([ADP][Pi])}. In brain, this increase in energy state stimulates dopaminergic neural activity signalling reward and a sense of well being, while suppressing glutamatergic neural activity signalling anxiety and unease. These positive responses to ethanol ingestion are important to social alcohol consumption. Importantly, decreased free [AMP] decreases AMP-dependent protein kinase (AMPK) activity, an important regulator of cellular energy metabolism. Oxidation of substrates used for energy metabolism in the absence of ethanol is down regulated to accommodate the input from ethanol. In liver, chronic ethanol metabolism results in fatty liver and general metabolic dysfunction. In brain, transport of other oxidizable metabolites through the blood-brain barrier and the enzymes for their oxidation are both down regulated. For exposures of short duration, ethanol induced regulatory changes are rapid and reversible, recovering completely when the concentrations of ethanol and acetate fall again. Longer periods of ethanol exposure and associated chronic suppression of AMPK activity activates regulatory mechanisms, including gene expression, that operate over longer time scales, both in onset and reversal. If chronic alcohol consumption is abruptly ended, metabolism is no longer able to respond rapidly enough to compensate. Glutamatergic neural activity adapts to chronic dysregulation of glutamate metabolism and suppression of glutamatergic neural activity by increasing excitatory and decreasing inhibitory amino acid receptors. A point is reached (ethanol dependence) where withdrawal of ethanol results in significant metabolic energy depletion in neurons and other brain cells as well as hyperexcitation of the glutamatergic system. The extent and regional specificity of energy depletion in the brain, combined with hyperactivity of the glutamatergic neuronal system, largely determines the severity of withdrawal symptoms.
Collapse
Affiliation(s)
- David F Wilson
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| | - Franz M Matschinsky
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
20
|
Jacob A, Wang P. Alcohol Intoxication and Cognition: Implications on Mechanisms and Therapeutic Strategies. Front Neurosci 2020; 14:102. [PMID: 32116535 PMCID: PMC7029710 DOI: 10.3389/fnins.2020.00102] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/27/2020] [Indexed: 12/23/2022] Open
Abstract
Binge alcohol drinking is highly prevalent in young adults and results in 30% deaths per year in young males. Binge alcohol drinking or acute alcohol intoxication is a risk factor for developing alcohol use disorder (AUD). Three FDA approved drugs are currently in use as therapy for AUD; however, all of them have contra-indications and limitations. Structural brain imaging studies in alcoholics have shown defects in the brain regions involved in memory, cognition and emotional processing. Positron emission tomography (PET) using radiotracers (e.g., 18FDG) and measuring brain glucose metabolism have demonstrated diagnostic and prognostic utility in evaluating patients with cognitive impairment. Using PET imaging, only a few exclusive human studies have addressed the relationship between alcohol intoxication and cognition. Those studies indicate that alcohol intoxication causes reduction in brain activity. Consistent with prior findings, a recent study by us showed that acute alcohol intoxication reduced brain activity in the cortical and subcortical regions including the temporal lobe consisting the hippocampus. Additionally, we have observed a strong correlation between reduction in metabolic activity and spatial cognition impairment in the hippocampus after binge alcohol exposure. We have also demonstrated the involvement of a stress response protein, cold inducible RNA binding protein (CIRP), as a potential mechanistic mediator in acute alcohol intoxication. In this review, we will first discuss in detail prior human PET imaging studies on alcohol intoxication as well as our recent study on acute alcohol intoxication, and review the existing literature on potential mechanisms of acute alcohol intoxication-induced cognitive impairment and therapeutic strategies to mitigate these impairments. Finally, we will highlight the importance of studying brain regions as part of a brain network in delineating the mechanism of acute alcohol intoxication-induced cognitive impairment to aid in the development of therapeutics for such indication.
Collapse
Affiliation(s)
- Asha Jacob
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
21
|
Abstract
Wine has historically been associated with religious rights, used as a salubrious beverage, employed as a medication as well as a medicinal solvent, and consumed as a food accompaniment. It is the last use that is most intimately associated in the minds of most modern consumers. Despite this, there is little flavor commonality on which pairing could be based. The first section of the chapter examines this feature and wine's primary role as a palate cleanser and food condiment. The synergistic role of food and wine in suppressing each other's least pleasant attributes is also explained. The final section deals with the latest evidence relating to the many beneficial health effects of moderate wine consumption, shortfalls in the data, headache induction, dental erosion, and conditions under which wine intake is contraindicated.
Collapse
|
22
|
Tanabe J, Yamamoto DJ, Sutton B, Brown MS, Hoffman PL, Burnham EL, Glueck DH, Tabakoff B. Effects of Alcohol and Acetate on Cerebral Blood Flow: A Pilot Study. Alcohol Clin Exp Res 2019; 43:2070-2078. [PMID: 31386214 PMCID: PMC7066986 DOI: 10.1111/acer.14173] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/25/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Acute alcohol produces effects on cerebral metabolism and blood flow. Alcohol is converted to acetate, which serves as a source of energy for the brain and is an agonist at G protein-coupled receptors distributed in different cell types in the body including neurons. Acetate has been hypothesized to play a role in the cerebral blood flow (CBF) response after alcohol ingestion. We tested whether administration of acetate would alter CBF in a pattern similar to or different from that of alcohol ingestion in healthy individuals. METHODS Twenty-four healthy participants were assigned by convenience to receive either 0.6 g/kg alcohol orally (n = 12) or acetate intravenously (n = 12). For each participant, CBF maps were acquired using an arterial spin labeling sequence on a 3T magnetic resonance scanner after placebo and after drug administration. Whole-brain CBF maps were compared between placebo and drug using a paired t-test, and set at a threshold of p < 0.05 corrected for multiple comparisons (k ≥ 142 voxels, ≥3.78 cm3 ), voxel-level p < 0.005. Intoxication was measured after placebo and drug administration with a Subjective High Assessment Scale (SHAS-7). RESULTS Compared to placebo, alcohol and acetate were associated with increased CBF in the medial thalamus. Alcohol, but not acetate, was associated with increased CBF in the right orbitofrontal, medial prefrontal and cingulate cortex, and hippocampus. Plasma acetate levels increased following administration of alcohol and acetate and did not differ between the 2 arms. Alcohol, but not acetate, was associated with an increase in SHAS-7 scores (p < 0.001). CONCLUSIONS Increased thalamic CBF associated with either alcohol or acetate administration suggests that the thalamic CBF response after alcohol could be mediated by acetate. Compared to other brain regions, thalamus may differ in its ability to metabolize acetate or expression of receptors responsive to acetate. Increased prefrontal and limbic CBF associated with alcohol may be linked to alcohol's behavioral effects.
Collapse
Affiliation(s)
- Jody Tanabe
- Department of Radiology, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Psychiatry, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dorothy J. Yamamoto
- Department of Radiology, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brianne Sutton
- Department of Radiology, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Psychiatry, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mark S. Brown
- Department of Radiology, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Paula L. Hoffman
- Department of Pharmacology, School of Medicine, University
of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ellen L. Burnham
- Department of Medicine, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Deborah H. Glueck
- Department of Pediatrics, School of Medicine, University of
Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Boris Tabakoff
- Department of Pharmaceutical Sciences, School of Pharmacy,
University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
23
|
Tomasi DG, Wiers CE, Shokri-Kojori E, Zehra A, Ramirez V, Freeman C, Burns J, Kure Liu C, Manza P, Kim SW, Wang GJ, Volkow ND. Association Between Reduced Brain Glucose Metabolism and Cortical Thickness in Alcoholics: Evidence of Neurotoxicity. Int J Neuropsychopharmacol 2019; 22:548-559. [PMID: 31369670 PMCID: PMC6754735 DOI: 10.1093/ijnp/pyz036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/12/2019] [Accepted: 07/15/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Excessive alcohol consumption is associated with reduced cortical thickness (CT) and lower cerebral metabolic rate of glucose (CMRGlu), but the correlation between these 2 measures has not been investigated. METHODS We tested the association between CT and cerebral CMRGlu in 19 participants with alcohol use disorder (AUD) and 20 healthy controls. Participants underwent 2-Deoxy-2-[18F]fluoroglucose positron emission tomography to map CMRGlu and magnetic resonance imaging to assess CT. RESULTS Although performance accuracy on a broad range of cognitive domains did not differ significantly between AUD and HC, AUD had widespread decreases in CT and CMRGlu. CMRGlu, normalized to cerebellum (rCMRGlu), showed significant correlation with CT across participants. Although there were large group differences in CMRGlu (>17%) and CT (>6%) in medial orbitofrontal and BA 47, the superior parietal cortex showed large reductions in CMRGlu (~17%) and minimal CT differences (~2.2%). Though total lifetime alcohol (TLA) was associated with CT and rCMRGlu, the causal mediation analysis revealed significant direct effects of TLA on rCMRGlu but not on CT, and there were no significant mediation effects of TLA, CT, and rCMRGlu. CONCLUSIONS The significant correlation between decrements in CT and CMRGlu across AUD participants is suggestive of alcohol-induced neurotoxicity, whereas the findings that the most metabolically affected regions in AUD had minimal atrophy and vice versa indicates that changes in CT and CMRGlu reflect distinct responses to alcohol across brain regions.
Collapse
Affiliation(s)
- Dardo G Tomasi
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD,Correspondence: Dardo Tomasi, PhD, 10 Center Dr, Rm B2L124, Bethesda, MD 20892-1013 ()
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | | | - Amna Zehra
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Veronica Ramirez
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Clara Freeman
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Jamie Burns
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | | | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Sung W Kim
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD,National Institute on Drug Abuse, Bethesda, MD
| |
Collapse
|
24
|
Abbasi Y, Shabani R, Mousavizadeh K, Soleimani M, Mehdizadeh M. Neuroprotective effect of ethanol and Modafinil on focal cerebral ischemia in rats. Metab Brain Dis 2019; 34:805-819. [PMID: 30644018 DOI: 10.1007/s11011-018-0378-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/26/2018] [Indexed: 01/08/2023]
Abstract
Ethanol is known as an effective agent against cerebral lesions after ischemia. Modafinil is a stimulant of the central nervous system (CNS) with antioxidant properties. We assessed the neuroprotective effect of modafinil in combination with ethanol after focal cerebral ischemia. Male wistar rats weighing 280-300 g were divided into nine groups (n = 12 each group): The groups consisted of the MCAO (middle cerebral artery occlusion) group (i.e. ischemia without treatment); the vehicle group(Dimethylsulfoxide); the modafinil group including three subgroups which pretreated with Modafinil (10, 30, 100 mg/kg), respectively, for seven days prior to the induction of MCAO; the ethanol group which received 1.5g/kg ethanol at the time of reperfusion; and modafinil+ethanol group which was divided into three subgroups that received three doses of modanifil (10, 30,100 mg/kg), respectively, for seven days prior to MCAO as well as ethanol at the time of reperfusion. Transient cerebral ischemia was induced by 60-min intraluminal occlusion of the right middle cerebral artery. Edema, infarct volume, glial scar formation (gliosis) and apoptosis were analyzed. The ethanol alone treatment (with a less significant effect), modafinil (in a dose-dependent way), and the combination of modafinil and ethanol significantly decreased the brain infarct volume, edema, apoptosis, and gliosis (P ≤ 0.05). Additionally, modafinil+ethanol mediated the restoration of aerobic metabolism and hyper-glycolysis suppress, thereby resulting in an increase in pyruvate dehydrogenase and a decrease in lactate dehydrogenase activity, respectively, which ultimately reduced oxidative reperfusion injury. These results demonstrate that pretreatment with modafinil (100 mg/kg) and modafinil+ethanol(1.5 g/kg) may prevent ischemic brain injuries.
Collapse
Affiliation(s)
- Yusef Abbasi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kazem Mousavizadeh
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Soleimani
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Mehdizadeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Jacob A, Ma Y, Nasiri E, Ochani M, Carrion J, Peng S, Brenner M, Huerta PT, Wang P. Extracellular cold inducible RNA-binding protein mediates binge alcohol-induced brain hypoactivity and impaired cognition in mice. Mol Med 2019; 25:24. [PMID: 31146675 PMCID: PMC6543653 DOI: 10.1186/s10020-019-0092-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/15/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alcohol abuse affects the brain regions responsible for memory, coordination and emotional processing. Binge alcohol drinking has shown reductions in brain activity, but the molecular targets have not been completely elucidated. We hypothesized that brain cells respond to excessive alcohol by releasing a novel inflammatory mediator, called cold inducible RNA-binding protein (CIRP), which is critical for the decreased brain metabolic activity and impaired cognition. METHODS Male wild type (WT) mice and mice deficient in CIRP (CIRP-/-) were studied before and after exposure to binge alcohol level by assessment of relative brain glucose metabolism with fluorodeoxyglucose (18FDG) and positron emission tomography (PET). Mice were also examined for object-place memory (OPM) and open field (OF) tasks. RESULTS Statistical Parametric Analysis (SPM) of 18FDG-PET uptake revealed marked decreases in relative glucose metabolism in distinct brain regions of WT mice after binge alcohol. Regional analysis (post hoc) revealed that while activity in the temporal (secondary visual) and limbic (entorhinal/perirhinal) cortices was decreased in WT mice, relative glucose metabolic activity was less suppressed in the CIRP-/- mice. Group and condition interaction analysis revealed differing responses in relative glucose metabolism (decrease in WT mice but increase in CIRP-/- mice) after alcohol in brain regions including the hippocampus and the cortical amygdala where the percent changes in metabolic activity correlated with changes in object discrimination performance. Behaviorally, alcohol-treated WT mice were impaired in exploring a repositioned object in the OPM task, and were more anxious in the OF task, whereas CIRP-/- mice were not impaired in these tasks. CONCLUSION CIRP released from brain cells could be responsible for regional brain metabolic hypoactivity leading to cognitive impairment under binge alcohol conditions.
Collapse
Affiliation(s)
- Asha Jacob
- Immunology and Inflammation, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Yilong Ma
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Elham Nasiri
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Mahendar Ochani
- Immunology and Inflammation, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Joseph Carrion
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Shichun Peng
- Center for Neurosciences, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
| | - Max Brenner
- Immunology and Inflammation, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Patricio T. Huerta
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| | - Ping Wang
- Immunology and Inflammation, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY USA
- Departments of Surgery and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY USA
| |
Collapse
|
26
|
Enculescu C, Kerr ED, Yeo KYB, Schenk G, Fortes MRS, Schulz BL. Proteomics Reveals Profound Metabolic Changes in the Alcohol Use Disorder Brain. ACS Chem Neurosci 2019; 10:2364-2373. [PMID: 30807102 DOI: 10.1021/acschemneuro.8b00660] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Changes in brain metabolism are a hallmark of alcohol use disorder (AUD). Determining how AUD changes the brain proteome is critical for understanding the effects of alcohol consumption on biochemical processes in the brain. We used data-independent acquisition mass spectrometry proteomics to study differences in the abundance of proteins associated with AUD in prefrontal lobe and motor cortex from autopsy brain. AUD had a substantial effect on the overall brain proteome exceeding the inherent differences between brain regions. Proteins associated with glycolysis, trafficking, the cytoskeleton, and excitotoxicity were altered in abundance in AUD. We observed extensive changes in the abundance of key metabolic enzymes, consistent with a switch from glucose to acetate utilization in the AUD brain. We propose that metabolic adaptations allowing efficient acetate utilization contribute to ethanol dependence in AUD.
Collapse
Affiliation(s)
- Charmaine Enculescu
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Edward D. Kerr
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - K. Y. Benjamin Yeo
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Gerhard Schenk
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Marina R. S. Fortes
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| | - Benjamin L. Schulz
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane 4072, Australia
| |
Collapse
|
27
|
Fritz M, Klawonn AM, Zahr NM. Neuroimaging in alcohol use disorder: From mouse to man. J Neurosci Res 2019; 100:1140-1158. [PMID: 31006907 DOI: 10.1002/jnr.24423] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/15/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
This article provides an overview of recent advances in understanding the effects of alcohol use disorders (AUD) on the brain from the perspective of magnetic resonance imaging (MRI) research in preclinical models and clinical studies. As a noninvasive investigational tool permitting assessment of morphological, metabolic, and hemodynamic changes over time, MRI offers insight into the dynamic course of alcoholism beginning with initial exposure through periods of binge drinking and escalation, sobriety, and relapse and has been useful in differential diagnosis of neurological diseases associated with AUD. Structural MRI has revealed acute and chronic effects of alcohol on both white and gray matter volumes. MR Spectroscopy, able to quantify brain metabolites in vivo, has shed light on biochemical alterations associated with alcoholism. Diffusion tensor imaging permits microstructural characterization of white matter fiber tracts. Functional MRI has allowed for elucidation of hemodynamic responses at rest and during task engagement. Positron emission tomography, a non-MRI imaging tool, has led to a deeper understanding of alcohol-induced receptor and neurotransmitter changes during various stages of drinking and abstinence. Together, such in vivo imaging tools have expanded our understanding of the dynamic course of alcoholism including evidence for regional specificity of the effects of AUD, hints at mechanisms underlying the shift from casual to compulsive use of alcohol, and profound recovery with sustained abstinence.
Collapse
Affiliation(s)
- Michael Fritz
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Natalie M Zahr
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California.,Neuroscience Program, SRI International, Menlo Park, California
| |
Collapse
|
28
|
Correspondence between cerebral glucose metabolism and BOLD reveals relative power and cost in human brain. Nat Commun 2019; 10:690. [PMID: 30741935 PMCID: PMC6370887 DOI: 10.1038/s41467-019-08546-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 01/17/2019] [Indexed: 12/02/2022] Open
Abstract
The correspondence between cerebral glucose metabolism (indexing energy utilization) and synchronous fluctuations in blood oxygenation (indexing neuronal activity) is relevant for neuronal specialization and is affected by brain disorders. Here, we define novel measures of relative power (rPWR, extent of concurrent energy utilization and activity) and relative cost (rCST, extent that energy utilization exceeds activity), derived from FDG-PET and fMRI. We show that resting-state networks have distinct energetic signatures and that brain could be classified into major bilateral segments based on rPWR and rCST. While medial-visual and default-mode networks have the highest rPWR, frontoparietal networks have the highest rCST. rPWR and rCST estimates are generalizable to other indexes of energy supply and neuronal activity, and are sensitive to neurocognitive effects of acute and chronic alcohol exposure. rPWR and rCST are informative metrics for characterizing brain pathology and alternative energy use, and may provide new multimodal biomarkers of neuropsychiatric disorders. The brain primarily uses glucose to generate energy, but the relationship of neuronal activity to glucose utilization is not necessarily a simple linear one. Here, the authors introduce relative power (rPWR) and relative cost (rCST) as new metrics to quantify how brain activity relates to glucose consumption.
Collapse
|
29
|
Dencker D, Molander A, Thomsen M, Schlumberger C, Wortwein G, Weikop P, Benveniste H, Volkow ND, Fink-Jensen A. Ketogenic Diet Suppresses Alcohol Withdrawal Syndrome in Rats. Alcohol Clin Exp Res 2017; 42:270-277. [PMID: 29160944 DOI: 10.1111/acer.13560] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Alcohol use disorder is underdiagnosed and undertreated, and up to 50% of alcohol-abstinent patients diagnosed with alcohol dependence relapse within the first year of treatment. Current treatments for the maintenance of alcohol abstinence in patients with alcohol use disorder have limited efficacy, and there is an urgent need for novel treatment strategies. Decreased cerebral glucose metabolism and increased brain uptake of acetate were recently reported in heavy drinkers, relative to controls. Given the switch of metabolic fuel from glucose to acetate in the alcohol-dependent brain, we investigated the potential therapeutic benefit of a ketogenic diet in managing alcohol withdrawal symptoms during detoxification. METHODS Male Sprague Dawley rats fed either ketogenic or regular diet were administered ethanol or water orally, twice daily for 6 days while the diet conditions were maintained. Abstinence symptoms were rated 6, 24, 48, and 72 hours after the last alcohol administration. RESULTS Maintenance on a ketogenic diet caused a significant decrease in the alcohol withdrawal symptoms' "rigidity" and "irritability." CONCLUSIONS Our preclinical pilot study suggests that a ketogenic diet may be a novel approach for treating alcohol withdrawal symptoms in humans.
Collapse
Affiliation(s)
- Ditte Dencker
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Anna Molander
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Morgane Thomsen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Chantal Schlumberger
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Gitta Wortwein
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Pia Weikop
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Helene Benveniste
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - Nora D Volkow
- Laboratory for Neuroimaging, National Institute on Alcohol Abuse and Alcoholism (NIAAA), Bethesda, Maryland
| | - Anders Fink-Jensen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
30
|
Hsieh YJ, Wu LC, Ke CC, Chang CW, Kuo JW, Huang WS, Chen FD, Yang BH, Tai HT, Chen SCJ, Liu RS. Effects of the Acute and Chronic Ethanol Intoxication on Acetate Metabolism and Kinetics in the Rat Brain. Alcohol Clin Exp Res 2017; 42:329-337. [PMID: 29205407 DOI: 10.1111/acer.13573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 11/20/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Ethanol (EtOH) intoxication inhibits glucose transport and decreases overall brain glucose metabolism; however, humans with long-term EtOH consumption were found to have a significant increase in [1-11 C]-acetate uptake in the brain. The relationship between the cause and effect of [1-11 C]-acetate kinetics and acute/chronic EtOH intoxication, however, is still unclear. METHODS [1-11 C]-acetate positron emission tomography (PET) with dynamic measurement of K1 and k2 rate constants was used to investigate the changes in acetate metabolism in different brain regions of rats with acute or chronic EtOH intoxication. RESULTS PET imaging demonstrated decreased [1-11 C]-acetate uptake in rat brain with acute EtOH intoxication, but this increased with chronic EtOH intoxication. Tracer uptake rate constant K1 and clearance rate constant k2 were decreased in acutely intoxicated rats. No significant change was noted in K1 and k2 in chronic EtOH intoxication, although 6 of 7 brain regions showed slightly higher k2 than baseline. These results indicate that acute EtOH intoxication accelerated acetate transport and metabolism in the rat brain, whereas chronic EtOH intoxication status showed no significant effect. CONCLUSIONS In vivo PET study confirmed the modulatory role of EtOH, administered acutely or chronically, in [1-11 C]-acetate kinetics and metabolism in the rat brain. Acute EtOH intoxication may inhibit the transport and metabolism of acetate in the brain, whereas chronic EtOH exposure may lead to the adaptation of the rat brain to EtOH in acetate utilization. [1-11 C]-acetate PET imaging is a feasible approach to study the effect of EtOH on acetate metabolism in rat brain.
Collapse
Affiliation(s)
- Ya-Ju Hsieh
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Liang-Chih Wu
- National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chien-Chih Ke
- Biomedical Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.,Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Chi-Wei Chang
- National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jung-Wen Kuo
- Biomedical Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.,Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Wen-Sheng Huang
- National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fu-Du Chen
- Biomedical Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Bang-Hung Yang
- National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Hsiao-Ting Tai
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Sharon Chia-Ju Chen
- Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ren-Shyan Liu
- National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Biomedical Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.,Molecular and Genetic Imaging Core/Taiwan Mouse Clinic, National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Biophotonic and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Nuclear Medicine, Cheng-Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
31
|
Forreider B, Pozivilko D, Kawaji Q, Geng X, Ding Y. Hibernation-like neuroprotection in stroke by attenuating brain metabolic dysfunction. Prog Neurobiol 2017; 157:174-187. [PMID: 26965388 DOI: 10.1016/j.pneurobio.2016.03.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 11/24/2022]
Abstract
Many mammalian species naturally undergo hibernation, a process that is associated with drastic changes in metabolism and systemic physiology. Their ability to retain an undamaged central nervous system during severely reduced cerebral blood flow has been studied for possible therapeutic application in human ischemic stroke. By inducing a less extreme 'hibernation-like' state, it has been hypothesized that similar neuroprotective effects reduce ischemia-mediated tissue damage in stroke patients. This manuscript includes reviews and evaluations of: (1) true hibernation, (2) hibernation-like state and its neuroprotective characteristics, (3) the preclinical and clinical methods for induction of artificial hibernation (i.e., therapeutic hypothermia, phenothiazine drugs, and ethanol), and (4) the mechanisms by which cerebral ischemia leads to tissue damage and how the above-mentioned induction methods function to inhibit those processes.
Collapse
Affiliation(s)
- Brian Forreider
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - David Pozivilko
- Michigan State University College of Human Medicine, East Lansing, MI, USA
| | - Qingwen Kawaji
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
| | - Yuchuan Ding
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
32
|
Alcohol affects brain functional connectivity and its coupling with behavior: greater effects in male heavy drinkers. Mol Psychiatry 2017; 22:1185-1195. [PMID: 27021821 PMCID: PMC5138152 DOI: 10.1038/mp.2016.25] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 02/02/2016] [Accepted: 02/04/2016] [Indexed: 01/14/2023]
Abstract
Acute and chronic alcohol exposure significantly affect behavior but the underlying neurobiological mechanisms are still poorly understood. Here, we used functional connectivity density (FCD) mapping to study alcohol-related changes in resting brain activity and their association with behavior. Heavy drinkers (HD, N=16, 16 males) and normal controls (NM, N=24, 14 males) were tested after placebo and after acute alcohol administration. Group comparisons showed that NM had higher FCD in visual and prefrontal cortices, default mode network regions and thalamus, while HD had higher FCD in cerebellum. Acute alcohol significantly increased FCD within the thalamus, impaired cognitive and motor functions, and affected self-reports of mood/drug effects in both groups. Partial least squares regression showed that alcohol-induced changes in mood/drug effects were associated with changes in thalamic FCD in both groups. Disruptions in motor function were associated with increases in cerebellar FCD in NM and thalamus FCD in HD. Alcohol-induced declines in cognitive performance were associated with connectivity increases in visual cortex and thalamus in NM, but in HD, increases in precuneus FCD were associated with improved cognitive performance. Acute alcohol reduced 'neurocognitive coupling', the association between behavioral performance and FCD (indexing brain activity), an effect that was accentuated in HD compared with NM. Findings suggest that reduced cortical connectivity in HD contribute to decline in cognitive abilities associated with heavy alcohol consumption, whereas increased cerebellar connectivity in HD may have compensatory effects on behavioral performance. The results reveal how drinking history alters the association between brain FCD and individual differences in behavioral performance.
Collapse
|
33
|
Haass-Koffler CL, Akhlaghi F, Swift RM, Leggio L. Altering ethanol pharmacokinetics to treat alcohol use disorder: Can you teach an old dog new tricks? J Psychopharmacol 2017; 31:812-818. [PMID: 28093021 PMCID: PMC5768306 DOI: 10.1177/0269881116684338] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Disulfiram was the first pharmacotherapy approved to treat alcohol use disorder in the 1950s. Disulfiram alters ethanol pharmacokinetics and causes uncomfortable reactions (e.g. headache, tachycardia, nausea, flushing and hypotension) when alcohol is consumed. Subsequently, a better understanding of the neurobiological pathways involved in alcohol use disorder led to the development of other medications (e.g. naltrexone and acamprosate). These neurobiological-based medications act on alcohol use disorder-related phenotypes including craving, stress, and/or withdrawal. The original approach to treat alcohol use disorder, by altering ethanol pharmacokinetics has been much less investigated. Recent research on ethanol pharmacokinetics has shed light on the mechanisms of action underlying alcohol use disorder and how some medications that alter ethanol pharmacokinetics may be helpful in treating alcohol use disorder. This review summarizes and discusses the complex pharmacokinetics of ethanol, and proposes that altering ethanol pharmacokinetics via novel pharmacological approaches may be a viable approach to treat alcohol use disorder.
Collapse
Affiliation(s)
- Carolina L. Haass-Koffler
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD
| | - Fatemeh Akhlaghi
- Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI
| | - Robert M. Swift
- Center for Alcohol and Addiction Studies, Department of Psychiatry and Human Behavior, Brown University, Providence, RI
- Veterans Affairs Medical Center, Providence, RI
| | - Lorenzo Leggio
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD
| |
Collapse
|
34
|
Effects of acute alcohol intoxication on executive functions controlling self-regulated behavior. Alcohol 2017; 61:1-8. [PMID: 28599712 DOI: 10.1016/j.alcohol.2017.02.177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 12/31/2016] [Accepted: 02/10/2017] [Indexed: 11/23/2022]
Abstract
Alcohol consumption may lead to deficits in the executive functions that govern self-regulation. These deficits could lead to risk-taking behaviors; therefore, it is important to determine the magnitude of these deficits on executive functioning. The purpose of this experiment was to investigate the acute effects of alcohol on three of the executive functions that are hypothesized to affect self-regulation, which are inhibition, set shifting, and working memory, using a mixed-methods study design. The participants were 75 moderate or heavy drinkers between the ages of 21 and 35 who were randomized into one of three beverage conditions (control, placebo, or 0.65-g alcohol dose/kg body weight). Performance on working memory, set shifting, and inhibition were measured pre- and post-beverage consumption. The results showed only a significant interaction in the working memory data, as there was an increase in performance post-beverage relative to pre-beverage for the control participants as compared to the alcohol and placebo participants. It was concluded that the dose of alcohol (BAC = 0.063%) given to moderate to heavy drinkers was not sufficient to cause significant impairment in the executive functions tested. The results were further discussed and methodological concerns were considered, such as the low BAC achieved, practice effects, and insensitivity of tasks.
Collapse
|
35
|
Gispert JD, Figueiras FP, Vengeliene V, Herance JR, Rojas S, Spanagel R. Changes in cerebral [ 18F]-FDG uptake induced by acute alcohol administration in a rat model of alcoholism. Behav Brain Res 2017; 327:29-33. [PMID: 28347826 DOI: 10.1016/j.bbr.2017.03.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 10/19/2022]
Abstract
Several [18F]-FDG positron emission tomography (PET) studies in alcoholics have consistently reported decreases in overall brain glucose metabolism at rest and following acute alcohol administration. However, changes in cerebral glucose utilization associated with the transition to addiction are not well understood and require longitudinal translational imaging studies in animal models of alcoholism. Here, we studied brain glucose uptake in alcohol drinking rats in order to provide convergent evidence to what has previously been reported in human studies. Brain glucose metabolism was measured by [18F]-FDG microPET imaging in different male Wistar rat groups: short-term drinking (three months), long-term drinking (twelve months) and alcohol-naïve. Global and regional cerebral glucose uptake was measured at rest and following acute alcohol administration. We showed that alcohol significantly reduced the whole-brain glucose metabolism. This effect was most pronounced in the parietal cortex and cerebellum. Alcohol-induced decreases in brain [18F]-FDG uptake was most apparent in alcohol-naïve rats, less intense in short-term drinkers and absent in long-term drinkers. The latter finding indicates the occurrence of tolerance to the intoxicating effects of alcohol in long-term drinking individuals. In contrast, some regions, like the ventral striatum and entorhinal cortex, showed enhanced metabolic activity, an effect that did not undergo tolerance during long-term alcohol consumption. Our findings are comparable to those described in human studies using the same methodology. We conclude that [18F]-FDG PET studies in rat models of alcoholism provide good translation and can be used for future longitudinal studies investigating alterations in brain function during different stages of the addiction cycle.
Collapse
Affiliation(s)
- Juan D Gispert
- Barcelonabeta Brain Research Center, Pasqual Maragall Foundation, Barcelona, Spain
| | - Francisca P Figueiras
- Institut d'Alta Tecnologia, Parc de Recerca Biomèdica de Barcelona (IAT- PRBB), CRC Corporació Sanitària, Barcelona, Spain
| | - Valentina Vengeliene
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - José R Herance
- Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Santiago Rojas
- Department of Morphological Sciences, Faculty of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| |
Collapse
|
36
|
Cui C, Koob GF. Titrating Tipsy Targets: The Neurobiology of Low-Dose Alcohol. Trends Pharmacol Sci 2017; 38:556-568. [PMID: 28372826 DOI: 10.1016/j.tips.2017.03.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/04/2017] [Accepted: 03/06/2017] [Indexed: 11/30/2022]
Abstract
Limited attention has been given to our understanding of how the brain responds to low-dose alcohol (ethanol) and what molecular and cellular targets mediate these effects. Even at concentrations lower than 10mM (0.046 g% blood alcohol concentration, BAC), below the legal driving limit in the USA (BAC 0.08 g%), alcohol impacts brain function and behavior. Understanding what molecular and cellular targets mediate the initial effects of alcohol and subsequent neuroplasticity could provide a better understanding of vulnerability or resilience to developing alcohol use disorders. We review here what is known about the neurobiology of low-dose alcohol, provide insights into potential molecular targets, and discuss future directions and challenges in further defining targets of low-dose alcohol at the molecular, cellular, and circuitry levels.
Collapse
Affiliation(s)
- Changhai Cui
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - George F Koob
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
37
|
Volkow ND, Wiers CE, Shokri-Kojori E, Tomasi D, Wang GJ, Baler R. Neurochemical and metabolic effects of acute and chronic alcohol in the human brain: Studies with positron emission tomography. Neuropharmacology 2017; 122:175-188. [PMID: 28108358 DOI: 10.1016/j.neuropharm.2017.01.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/20/2016] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
The use of Positron emission tomography (PET) to study the effects of acute and chronic alcohol on the human brain has enhanced our understanding of the mechanisms underlying alcohol's rewarding effects, the neuroadaptations from chronic exposure that contribute to tolerance and withdrawal, and the changes in fronto-striatal circuits that lead to loss of control and enhanced motivation to drink that characterize alcohol use disorders (AUD). These include studies showing that alcohol's reinforcing effects may result not only from its enhancement of dopaminergic, GABAergic and opioid signaling but also from its caloric properties. Studies in those suffering from an AUD have revealed significant alterations in dopamine (DA), GABA, cannabinoids, opioid and serotonin neurotransmission and in brain energy utilization (glucose and acetate metabolism) that are likely to contribute to compulsive alcohol taking, dysphoria/depression, and to alcohol-associated neurotoxicity. Studies have also evaluated the effects of abstinence on recovery of brain metabolism and neurotransmitter function and the potential value of some of these measures to predict clinical outcomes. Finally, PET studies have started to provide insights about the neuronal mechanisms by which certain genes contribute to the vulnerability to AUD. These findings have helped identify new strategies for prevention and treatment of AUD. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Nora D Volkow
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, United States; National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892, United States.
| | - Corinde E Wiers
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ehsan Shokri-Kojori
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, United States
| | - Dardo Tomasi
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, United States
| | - Gene-Jack Wang
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20892, United States
| | - Ruben Baler
- National Institute on Alcohol Abuse and Alcoholism, Laboratory of Neuroimaging, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
38
|
Phillips JG, Ogeil RP. Cannabis, alcohol use, psychological distress, and decision-making style. J Clin Exp Neuropsychol 2016; 39:670-681. [PMID: 27876440 DOI: 10.1080/13803395.2016.1255311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION There have been suggestions of hypofrontality in cannabis users. To understand cannabis-related differences in decisional processes, Janis and Mann's conflict model of decision making was applied to recreational cannabis smokers who varied in their alcohol use and level of psychological distress. METHOD An online sample of recreational substance users (114 male, 119 female) completed the Melbourne Decision Making Questionnaire, the Alcohol Use Disorders Identification Test (AUDIT), Kessler's Psychological Distress Scale (K10), and the Severity of Dependence Scale (SDS) for cannabis. RESULTS Multivariate analysis of variance examined self-reported decision-making styles as a function of gender, recent cannabis use, risky alcohol consumption, and levels of psychological distress. Psychological distress was associated with lower decisional self-esteem and higher levels of procrastination and buck-passing. There were gender differences associated with cannabis use. Female cannabis users reported higher levels of hypervigilance, while male cannabis users reported lower levels of buck-passing. CONCLUSIONS Although there was little indication of an avoidant decisional style in cannabis users, the results suggest that cannabis affects decisional processes, contributing to panic in females and impulsivity in males.
Collapse
Affiliation(s)
- James G Phillips
- a Psychology Department , Auckland University of Technology , Auckland , New Zealand
| | - Rowan P Ogeil
- b Eastern Health Clinical School , Monash University, and Turning Point, Eastern Health , Clayton , Australia
| |
Collapse
|
39
|
Nixon SJ, Prather R, Lewis B. Sex differences in alcohol-related neurobehavioral consequences. HANDBOOK OF CLINICAL NEUROLOGY 2016; 125:253-72. [PMID: 25307580 DOI: 10.1016/b978-0-444-62619-6.00016-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
In this chapter, we review existing research regarding sex differences in alcohol's effects on neurobehavioral functions/processes. Drawn largely from laboratory studies, literature regarding acute alcohol administration and chronic alcohol misuse is explored focusing on commonly employed neuropsychologic domains (e.g., executive function, visuospatial skills, learning and memory, gait and balance), neurophysiologic measures (e.g., electroencephalography and event-related potentials), and structural and functional neuroimaging (e.g., magnetic resonance imaging (MRI), functional MRI, diffusion tensor imaging, positron emission tomography, and magnetic resonance spectroscopy). To provide a historical perspective on the development of these questions, we have included reference to early and more recent research. Additionally, specific biases, knowledge gaps, and continuing controversies are noted.
Collapse
Affiliation(s)
- Sara Jo Nixon
- Department of Psychiatry, University of Florida, Gainesville, FL, USA; Department of Psychology, University of Florida, Gainesville, FL, USA.
| | - Robert Prather
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| | - Ben Lewis
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
| |
Collapse
|
40
|
Daulatzai MA. Cerebral hypoperfusion and glucose hypometabolism: Key pathophysiological modulators promote neurodegeneration, cognitive impairment, and Alzheimer's disease. J Neurosci Res 2016; 95:943-972. [PMID: 27350397 DOI: 10.1002/jnr.23777] [Citation(s) in RCA: 304] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 02/06/2023]
Abstract
Aging, hypertension, diabetes, hypoxia/obstructive sleep apnea (OSA), obesity, vitamin B12/folate deficiency, depression, and traumatic brain injury synergistically promote diverse pathological mechanisms including cerebral hypoperfusion and glucose hypometabolism. These risk factors trigger neuroinflammation and oxidative-nitrosative stress that in turn decrease nitric oxide and enhance endothelin, Amyloid-β deposition, cerebral amyloid angiopathy, and blood-brain barrier disruption. Proinflammatory cytokines, endothelin-1, and oxidative-nitrosative stress trigger several pathological feedforward and feedback loops. These upstream factors persist in the brain for decades, upregulating amyloid and tau, before the cognitive decline. These cascades lead to neuronal Ca2+ increase, neurodegeneration, cognitive/memory decline, and Alzheimer's disease (AD). However, strategies are available to attenuate cerebral hypoperfusion and glucose hypometabolism and ameliorate cognitive decline. AD is the leading cause of dementia among the elderly. There is significant evidence that pathways involving inflammation and oxidative-nitrosative stress (ONS) play a key pathophysiological role in promoting cognitive dysfunction. Aging and several comorbid conditions mentioned above promote diverse pathologies. These include inflammation, ONS, hypoperfusion, and hypometabolism in the brain. In AD, chronic cerebral hypoperfusion and glucose hypometabolism precede decades before the cognitive decline. These comorbid disease conditions may share and synergistically activate these pathophysiological pathways. Inflammation upregulates cerebrovascular pathology through proinflammatory cytokines, endothelin-1, and nitric oxide (NO). Inflammation-triggered ONS promotes long-term damage involving fatty acids, proteins, DNA, and mitochondria; these amplify and perpetuate several feedforward and feedback pathological loops. The latter includes dysfunctional energy metabolism (compromised mitochondrial ATP production), amyloid-β generation, endothelial dysfunction, and blood-brain-barrier disruption. These lead to decreased cerebral blood flow and chronic cerebral hypoperfusion- that would modulate metabolic dysfunction and neurodegeneration. In essence, hypoperfusion deprives the brain from its two paramount trophic substances, viz., oxygen and nutrients. Consequently, the brain suffers from synaptic dysfunction and neuronal degeneration/loss, leading to both gray and white matter atrophy, cognitive dysfunction, and AD. This Review underscores the importance of treating the above-mentioned comorbid disease conditions to attenuate inflammation and ONS and ameliorate decreased cerebral blood flow and hypometabolism. Additionally, several strategies are described here to control chronic hypoperfusion of the brain and enhance cognition. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mak Adam Daulatzai
- Sleep Disorders Group, EEE Dept/MSE, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
41
|
Cai L, Stevenson J, Peng C, Xin R, Rastogi R, Liu K, Geng X, Gao Z, Ji X, Rafols JA, Ji Z, Ding Y. Adjuvant therapies using normobaric oxygen with hypothermia or ethanol for reducing hyperglycolysis in thromboembolic cerebral ischemia. Neuroscience 2016; 318:45-57. [PMID: 26794589 DOI: 10.1016/j.neuroscience.2016.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/25/2015] [Accepted: 01/06/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND PURPOSE Normobaric oxygen (NBO), ethanol (EtOH), and therapeutic hypothermia (TH) delivered alone or in combination have neuroprotective properties after acute stroke. We used an autologous thromboembolic rat stroke model to assess the additive effects of these treatments for reducing the deleterious effects of hyperglycolysis post-stroke in which reperfusion is induced with recombinant tissue plasminogen activator (rt-PA). METHODS Sprague-Dawley rats were subjected to middle cerebral artery (MCA) occlusion with an autologous embolus. One hour after occlusion, rt-PA was administered alone or with NBO (60%), EtOH (1.0 g/kg), TH (33 °C), either singly or in combination. Infarct volume and neurological deficit were assessed at 24h after rt-PA-induced reperfusion with or without other treatments. The extent of hyperglycolysis, as determined by cerebral glucose and lactate levels was evaluated at 3 and 24h after rt-PA administration. At the same time points, expressions of glucose transporter 1 (Glut1), glucose transporter 3 (Glut3), phosphofructokinase1 (PFK-1), and lactate dehydrogenase were (LDH) measured by Western blotting. RESULTS Following rt-PA in rats with thromboembolic stroke, NBO combined with TH or EtOH most effectively decreased infarct volume and neurological deficit. As compared to rt-PA alone, EtOH or TH but not NBO monotherapies significantly reduced post-stroke hyperglycolysis. The increased utilization of glucose and production of lactate post-stroke was prevented most effectively when NBO was combined with either EtOH or TH after reperfusion with rt-PA, as shown by the significantly decreased Glut1, Glut3, PFK-1, and LDH levels. CONCLUSIONS In a rat thromboembolic stroke model, both EtOH and TH used individually offer neuroprotection after the administration of rt-PA. While NBO monotherapy does not appear to be effective, it significantly potentiates the efficacy of EtOH and TH. The similar neuroprotection and underlying mechanisms pertaining to the attenuation of hyperglycolysis provided by EtOH or TH in combination with NBO suggest a possibility of substituting EtOH for TH. Thus a combination of NBO and EtOH, which are widely available and easily used, could become a novel and effective neuroprotective strategy in the clinical setting.
Collapse
Affiliation(s)
- L Cai
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - J Stevenson
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - C Peng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - R Xin
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Radiology, Luhe Hospital, Capital Medical University, Beijing, China
| | - R Rastogi
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - K Liu
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - X Geng
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Z Gao
- Cerebral Vascular Diseases Research Institute, Capital Medical University, Beijing, China
| | - X Ji
- Cerebral Vascular Diseases Research Institute, Capital Medical University, Beijing, China
| | - J A Rafols
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Z Ji
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China.
| | - Y Ding
- China-America Institute of Neuroscience, Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
42
|
Song BJ, Akbar M, Jo I, Hardwick JP, Abdelmegeed MA. Translational Implications of the Alcohol-Metabolizing Enzymes, Including Cytochrome P450-2E1, in Alcoholic and Nonalcoholic Liver Disease. ADVANCES IN PHARMACOLOGY 2015; 74:303-72. [PMID: 26233911 DOI: 10.1016/bs.apha.2015.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fat accumulation (hepatic steatosis) in alcoholic and nonalcoholic fatty liver disease is a potentially pathologic condition which can progress to steatohepatitis (inflammation), fibrosis, cirrhosis, and carcinogenesis. Many clinically used drugs or some alternative medicine compounds are also known to cause drug-induced liver injury, which can further lead to fulminant liver failure and acute deaths in extreme cases. During liver disease process, certain cytochromes P450 such as the ethanol-inducible cytochrome P450-2E1 (CYP2E1) and CYP4A isozymes can be induced and/or activated by alcohol and/or high-fat diets and pathophysiological conditions such as fasting, obesity, and diabetes. Activation of these P450 isozymes, involved in the metabolism of ethanol, fatty acids, and various drugs, can produce reactive oxygen/nitrogen species directly and/or indirectly, contributing to oxidative modifications of DNA/RNA, proteins and lipids. In addition, aldehyde dehydrogenases including the mitochondrial low Km aldehyde dehydrogenase-2 (ALDH2), responsible for the metabolism of acetaldehyde and lipid aldehydes, can be inactivated by various hepatotoxic agents. These highly reactive acetaldehyde and lipid peroxides, accumulated due to ALDH2 suppression, can interact with cellular macromolecules DNA/RNA, lipids, and proteins, leading to suppression of their normal function, contributing to DNA mutations, endoplasmic reticulum stress, mitochondrial dysfunction, steatosis, and cell death. In this chapter, we specifically review the roles of the alcohol-metabolizing enzymes including the alcohol dehydrogenase, ALDH2, CYP2E1, and other enzymes in promoting liver disease. We also discuss translational research opportunities with natural and/or synthetic antioxidants, which can prevent or delay the onset of inflammation and liver disease.
Collapse
Affiliation(s)
- Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | - Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, South Korea
| | - James P Hardwick
- Biochemistry and Molecular Pathology in Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Daulatzai MA. “Boomerang Neuropathology” of Late-Onset Alzheimer’s Disease is Shrouded in Harmful “BDDS”: Breathing, Diet, Drinking, and Sleep During Aging. Neurotox Res 2015; 28:55-93. [PMID: 25911292 DOI: 10.1007/s12640-015-9528-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 04/03/2015] [Accepted: 04/03/2015] [Indexed: 12/12/2022]
|
44
|
Alcohol decreases baseline brain glucose metabolism more in heavy drinkers than controls but has no effect on stimulation-induced metabolic increases. J Neurosci 2015; 35:3248-55. [PMID: 25698759 DOI: 10.1523/jneurosci.4877-14.2015] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
During alcohol intoxication, the human brain increases metabolism of acetate and decreases metabolism of glucose as energy substrate. Here we hypothesized that chronic heavy drinking facilitates this energy substrate shift both for baseline and stimulation conditions. To test this hypothesis, we compared the effects of alcohol intoxication (0.75 g/kg alcohol vs placebo) on brain glucose metabolism during video stimulation (VS) versus when given with no stimulation (NS), in 25 heavy drinkers (HDs) and 23 healthy controls, each of whom underwent four PET-(18)FDG scans. We showed that resting whole-brain glucose metabolism (placebo-NS) was lower in HD than controls (13%, p = 0.04); that alcohol (compared with placebo) decreased metabolism more in HD (20 ± 13%) than controls (9 ± 11%, p = 0.005) and in proportion to daily alcohol consumption (r = 0.36, p = 0.01) but found that alcohol did not reduce the metabolic increases in visual cortex from VS in either group. Instead, VS reduced alcohol-induced decreases in whole-brain glucose metabolism (10 ± 12%) compared with NS in both groups (15 ± 13%, p = 0.04), consistent with stimulation-related glucose metabolism enhancement. These findings corroborate our hypothesis that heavy alcohol consumption facilitates use of alternative energy substrates (i.e., acetate) for resting activity during intoxication, which might persist through early sobriety, but indicate that glucose is still favored as energy substrate during brain stimulation. Our findings are consistent with reduced reliance on glucose as the main energy substrate for resting brain metabolism during intoxication (presumably shifting to acetate or other ketones) and a priming of this shift in HDs, which might make them vulnerable to energy deficits during withdrawal.
Collapse
|
45
|
The neurometabolic fingerprint of excessive alcohol drinking. Neuropsychopharmacology 2015; 40:1259-68. [PMID: 25418809 PMCID: PMC4367471 DOI: 10.1038/npp.2014.312] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/28/2014] [Accepted: 11/13/2014] [Indexed: 01/03/2023]
Abstract
'Omics' techniques are widely used to identify novel mechanisms underlying brain function and pathology. Here we applied a novel metabolomics approach to further ascertain the role of frontostriatal brain regions for the expression of addiction-like behaviors in rat models of alcoholism. Rats were made alcohol dependent via chronic intermittent alcohol vapor exposure. Following a 3-week abstinence period, rats had continuous access to alcohol in a two-bottle, free-choice paradigm for 7 weeks. Nontargeted flow injection time-of-flight mass spectrometry was used to assess global metabolic profiles of two cortical (prelimbic and infralimbic) and two striatal (accumbens core and shell) brain regions. Alcohol consumption produces pronounced global effects on neurometabolomic profiles leading to a clear separation of metabolic phenotypes between treatment groups, particularly. Further comparisons of regional tissue levels of various metabolites, most notably dopamine and Met-enkephalin, allow the extrapolation of alcohol consumption history. Finally, a high-drinking metabolic fingerprint was identified indicating a distinct alteration of central energy metabolism in the accumbens shell of excessively drinking rats that could indicate a so far unrecognized pathophysiological mechanism in alcohol addiction. In conclusion, global metabolic profiling from distinct brain regions by mass spectrometry identifies profiles reflective of an animal's drinking history and provides a versatile tool to further investigate pathophysiological mechanisms in alcohol dependence.
Collapse
|
46
|
Brain: normal variations and benign findings in fluorodeoxyglucose-PET/computed tomography imaging. PET Clin 2015; 9:129-40. [PMID: 24772054 DOI: 10.1016/j.cpet.2013.10.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Brain 18F-fluorodeoxyglucose (18F-FDG) PET allows the in vivo study of cerebral glucose metabolism, reflecting neuronal and synaptic activity. 18F-FDG-PET has been extensively used to detect metabolic alterations in several neurologic diseases compared with normal aging. However, healthy subjects have variants of 18F-FDG distribution, especially as associated with aging. This article focuses on 18F-FDG-PET findings in so-called normal brain aging, and in particular on metabolic differences occurring with aging and as a function of people’s gender. The effect of different substances, medications, and therapy procedures are discussed, as well as common artifacts.
Collapse
|
47
|
Bjork JM, Gilman JM. The effects of acute alcohol administration on the human brain: insights from neuroimaging. Neuropharmacology 2014; 84:101-10. [PMID: 23978384 PMCID: PMC3971012 DOI: 10.1016/j.neuropharm.2013.07.039] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 07/26/2013] [Accepted: 07/29/2013] [Indexed: 02/05/2023]
Abstract
Over the last quarter century, researchers have peered into the living human brain to develop and refine mechanistic accounts of alcohol-induced behavior, as well as neurobiological mechanisms for development and maintenance of addiction. These in vivo neuroimaging studies generally show that acute alcohol administration affects brain structures implicated in motivation and behavior control, and that chronic intoxication is correlated with structural and functional abnormalities in these same structures, where some elements of these decrements normalize with extended sobriety. In this review, we will summarize recent findings about acute human brain responses to alcohol using neuroimaging techniques, and how they might explain behavioral effects of alcohol intoxication. We then briefly address how chronic alcohol intoxication (as inferred from cross-sectional differences between various drinking populations and controls) may yield individual brain differences between drinking subjects that may confound interpretation of acute alcohol administration effects. This article is part of the Special Issue Section entitled 'Neuroimaging in Neuropharmacology'.
Collapse
Affiliation(s)
- James M Bjork
- Division of Clinical Neuroscience and Behavioral Research, National Institute on Drug Abuse, National Institutes of Health, 6001 Executive Blvd, Room 3163, Bethesda, MD 20892, USA.
| | - Jodi M Gilman
- Laboratory of Neuroimaging and Genetics, MGH Division of Psychiatric Neuroscience, Martinos Center for Biomedical Imaging, Massachusetts General Hospital, USA
| |
Collapse
|
48
|
Abstract
Neuroimaging, including PET, MRI, and MRS, is a powerful approach to the study of brain function. This article reviews neuroimaging findings related to alcohol and other drugs of abuse that have been published since 2011. Uses of neuroimaging are to characterize patients to determine who will fare better in treatment and to investigate the reasons underlying the effect on outcomes. Neuroimaging is also used to characterize the acute and chronic effects of substances on the brain and how those effects are related to dependence, relapse, and other drug effects. The data can be used to provide encouraging information for patients, as several studies have shown that long-term abstinence is associated with at least partial normalization of neurological abnormalities.
Collapse
Affiliation(s)
- Mark J Niciu
- National Institutes of Health and Department of Health and Human Services, Experimental Therapeutics & Pathophysiology Branch, National Institute of Mental Health, 10 Center Dr., Building 10/CRC, Room 7-5545, Bethesda, MD 20892, USA
| | - Graeme F Mason
- Yale University Department of Diagnostic Radiology and Psychiatry, New Haven, CT, USA
| |
Collapse
|
49
|
Sharma S, Nepal B, Moon CS, Chabenne A, Khogali A, Ojo C, Hong E, Gaudet R, Sayed-Ahmad A, Jacob A, Murtuza M, Firlit M. Psychology of Craving. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/ojmp.2014.32015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
50
|
Rae CD, Davidson JE, Maher AD, Rowlands BD, Kashem MA, Nasrallah FA, Rallapalli SK, Cook JM, Balcar VJ. Ethanol, not detectably metabolized in brain, significantly reduces brain metabolism, probably via action at specific GABA(A) receptors and has measureable metabolic effects at very low concentrations. J Neurochem 2013; 129:304-14. [PMID: 24313287 DOI: 10.1111/jnc.12634] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 11/12/2013] [Accepted: 12/04/2013] [Indexed: 11/28/2022]
Abstract
Ethanol is a known neuromodulatory agent with reported actions at a range of neurotransmitter receptors. Here, we measured the effect of alcohol on metabolism of [3-¹³C]pyruvate in the adult Guinea pig brain cortical tissue slice and compared the outcomes to those from a library of ligands active in the GABAergic system as well as studying the metabolic fate of [1,2-¹³C]ethanol. Analyses of metabolic profile clusters suggest that the significant reductions in metabolism induced by ethanol (10, 30 and 60 mM) are via action at neurotransmitter receptors, particularly α4β3δ receptors, whereas very low concentrations of ethanol may produce metabolic responses owing to release of GABA via GABA transporter 1 (GAT1) and the subsequent interaction of this GABA with local α5- or α1-containing GABA(A)R. There was no measureable metabolism of [1,2-¹³C]ethanol with no significant incorporation of ¹³C from [1,2-¹³C]ethanol into any measured metabolite above natural abundance, although there were measurable effects on total metabolite sizes similar to those seen with unlabelled ethanol.
Collapse
Affiliation(s)
- Caroline D Rae
- Neuroscience Research Australia, and Brain Sciences UNSW, Randwick, NSW, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|