201
|
Lu L, Yang W, Zhao D, Wen X, Liu J, Liu J, Yuan K. Brain recovery of the NAc fibers and prediction of craving changes in person with heroin addiction: A longitudinal study. Drug Alcohol Depend 2023; 243:109749. [PMID: 36565569 DOI: 10.1016/j.drugalcdep.2022.109749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/14/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Progress have been made in brain function recovery after long-term abstinence in person with heroin addiction (PHA). However, less is known about whether the nucleus accumbens (NAc) white matter pathways can recover in PHA by prolonged abstinence. METHODS Forty-two PHA and Thirty-nine age- and gender- matched healthy controls (HCs) were recruited. Two MRI scans were obtained at baseline (PHA1) and 8-month follow-up (PHA2). We employed tractography atlas-based analysis (TABS) method to investigate fractional anisotropy (FA) changes in NAc fiber tracts (i.e., Insula-NAc, ventral tegmental area (VTA)-NAc, medial prefrontal cortex (MPFC)-NAc) in PHA. A partial least square regression (PLSR) analysis was carried to explore whether FA of NAc fiber tracts can predict longitudinal craving changes. RESULTS Relative to HCs, lower FA was found in the right Insula-NAc and VTA-NAc fiber tracts in PHA1, and PHA2 showed increased FA values in these tracts compared with PHA1. Furthermore, changes of FA of NAc fiber tracts can predict longitudinal craving changes (r = 0.51). Additionally, craving changes can also be predicted from FA changes in the left Insula-NAc (r = 0.601) and VTA-NAc (r = 0.384) fiber alone. CONCLUSIONS Results indicated that the right Insula-NAc and VTA-NAc fiber tracts are potential biomarkers for brain recovery. Prediction of craving changes highlighted the utility of structural markers to inform clinical decision-making of treatment for PHA.
Collapse
Affiliation(s)
- Ling Lu
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Wenhan Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Desheng Zhao
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Xinwen Wen
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China.
| | - Jixin Liu
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, Shaanxi, 710071, China.
| | - Kai Yuan
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Engineering Research Center of Molecular and Neuro Imaging Ministry of Education, Xi'an, Shaanxi, 710071, China; International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China; Information Processing Laboratory, School of Information Engineering, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, 014010, China.
| |
Collapse
|
202
|
Berg M, Riehle M, Rief W, Lincoln T. Does partial blockade of dopamine D2 receptors with Amisulpride cause anhedonia? An experimental study in healthy volunteers. J Psychiatr Res 2023; 158:409-416. [PMID: 36680855 DOI: 10.1016/j.jpsychires.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/22/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
BACKGROUND Anhedonia is a frequent cause of functional impairment in psychosis. Although it is plausible that medication-induced D2 receptor blockade could diminish hedonic responding, there is little experimental research testing this hypothesis in humans. METHODS To inspect possible effects of partial D2 blockade on hedonic experiences, we administered 300 mg of Amisulpride or placebo to 85 participants in a randomized, double-blind, placebo-controlled trial. Participants were then subjected to an emotional evocation task utilizing standardized pictorial pleasant, neutral, and unpleasant stimuli. RESULTS We observed lower positivity ratings in the Amisulpride group compared to placebo across all stimulus categories (p = .026, f = 0.25) and no group differences in negativity or arousal ratings. The Amisulpride group also showed lower electrodermal responses across all stimulus categories compared to placebo (p = .017, f = 0.27). The electrodermal response was especially diminished for pleasant stimuli. CONCLUSION We interpret our findings as evidence that D2 blockade via Amisulpride can reduce at-the-moment hedonic responsivity in healthy volunteers. If these results can be confirmed in drug-naïve clinical samples, this would indicate that antipsychotic medication contributes to clinical anhedonia, probably via antagonistic effects at the dopamine D2 receptor.
Collapse
Affiliation(s)
- Max Berg
- Philipps-University of Marburg, Dept. of Psychology, Division of Clinical Psychology and Psychotherapy, Gutenbergstraße 18, D-35032, Marburg, Germany.
| | - Marcel Riehle
- Universität Hamburg, Dept. of Psychology, Clinical Psychology and Psychotherapy, Von-Melle-Park 5, D-20146, Hamburg, Germany
| | - Winfried Rief
- Philipps-University of Marburg, Dept. of Psychology, Division of Clinical Psychology and Psychotherapy, Gutenbergstraße 18, D-35032, Marburg, Germany
| | - Tania Lincoln
- Universität Hamburg, Dept. of Psychology, Clinical Psychology and Psychotherapy, Von-Melle-Park 5, D-20146, Hamburg, Germany
| |
Collapse
|
203
|
Ou CY, Yu YH, Wu CW, Kozłowska A, Shyu BC, Huang ACW. Neuronal activity of the medial prefrontal cortex, nucleus accumbens, and basolateral amygdala in conditioned taste aversion and conditioned place preference induced by different doses of morphine administrations in rats. Front Pharmacol 2023; 14:1062169. [PMID: 36762112 PMCID: PMC9902353 DOI: 10.3389/fphar.2023.1062169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
To re-examine the paradoxical effect hypothesis of abused drugs, the present study concerned whether different doses of morphine disparately affect neuronal activity and associations among the subareas of the medial prefrontal cortex (mPFC: cingulate cortex 1-Cg1, prelimbic cortex-PrL, infralimbic cortex-IL), the subregions of the nucleus accumbens (NAc; both core and shell), and the basolateral amygdala (BLA) following conditioned taste aversion (CTA) and conditioned place preference (CPP). All rats were given a 0.1% saccharin solution for 15-min, and they were intraperitoneally injected with saline or 20, 30, or 40 mg/kg morphine to form the aversive CTA learning. Later, half of the rats were tested for CPP (including the CTA and then CPP tests) for 30-min. Finally, the immunohistochemical staining with c-Fos was conducted after the behavioral test. After the CTA test, c-Fos (%) in the Cg1 and PrL (but not the IL) was more in 20-40 mg/kg of the morphine groups; c-Fos (%) in the NAc core, NAc shell, and BLA was more in the 30-40 mg/kg morphine group. After the CPP test, the Cg1, PrL, IL, and BLA showed more c-Fos (%) in 20 mg/kg morphine; the NAc core showed fewer in c-Fos (%) in the 30-40 mg/kg morphine groups. The mPFC subregions (e.g., Cg1, PrL, and IL), NAc subareas (e.g., NAc core and NAc shell), and BLA were involved in the different doses of morphine injections. The correlation analysis showed that a positive correlation was observed between PrL and IL with NAc core with low doses of morphine and with NAc shell with increasing doses of morphine after the CTA test. After the CPP, an association between PrL and NAc core and NAc shell at low doses and between IL and BLA and NAc shell with increasing doses of morphine. Therefore, different neural substrates and the neural connectivity are observed following different doses of morphine and after the CTA and CPP tests. The present data extend the paradoxical effect hypothesis of abused drugs.
Collapse
Affiliation(s)
- Chen Yin Ou
- Department of Psychology, Fo GuangUniversity, Jiaoxi, Yilan County, Taiwan
| | - Ying Hao Yu
- Department of Psychology, Fo GuangUniversity, Jiaoxi, Yilan County, Taiwan,Department of Biotechnology and Animal Science, National ILan University, Yilan, Taiwan
| | - Chi-Wen Wu
- Department of Psychology, Fo GuangUniversity, Jiaoxi, Yilan County, Taiwan,Department of Pharmacy, Keelung Hospital, Ministry of Health and Welfare, Keelung City, Taiwan
| | - Anna Kozłowska
- Department of Human Physiology and Pathology, School Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Bai-Chung Shyu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan,*Correspondence: Bai-Chung Shyu, ; Andrew Chih Wei Huang,
| | - Andrew Chih Wei Huang
- Department of Psychology, Fo GuangUniversity, Jiaoxi, Yilan County, Taiwan,*Correspondence: Bai-Chung Shyu, ; Andrew Chih Wei Huang,
| |
Collapse
|
204
|
Wiaderkiewicz J, Reilly S. Expression of c-Fos following voluntary ingestion of a novel or familiar taste in rats. Brain Res 2023; 1799:148177. [PMID: 36503889 PMCID: PMC9795852 DOI: 10.1016/j.brainres.2022.148177] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/28/2022]
Abstract
Taste neophobia, the rejection of novel tastes or foods, involves an interplay of various brain regions encompassing areas within the central gustatory system, as well as nuclei serving other functions. Previous findings, utilising c-Fos imaging, identified several brain regions which displayed higher activity after ingestion of a novel taste as compared to a familiar taste. The present study extends this analysis to include additional regions suspected of contributing to the neurocircuitry involved in evoking taste neophobia. Our data show increased c-Fos expression in the basolateral amygdala, central nucleus of the amygdala, gustatory portion of the thalamus, gustatory portion of the insular cortex and the medial and lateral regions of the parabrachial nucleus. These results confirm the contribution of areas previously identified as active during ingestion of novel tastes and expose additional areas that express elevated levels of c-Fos under these conditions, thus adding to the neural network involved in the detection and initial processing of taste novelty.
Collapse
Affiliation(s)
- Jan Wiaderkiewicz
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, United States.
| | - Steve Reilly
- Department of Psychology, University of Illinois at Chicago, 1007 West Harrison Street, Chicago, IL 60607, United States.
| |
Collapse
|
205
|
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
|
206
|
Jin D, Chen H, Chen SR, Pan HL. α2δ-1 protein drives opioid-induced conditioned reward and synaptic NMDA receptor hyperactivity in the nucleus accumbens. J Neurochem 2023; 164:143-157. [PMID: 36222452 PMCID: PMC9892208 DOI: 10.1111/jnc.15706] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/19/2022] [Accepted: 10/08/2022] [Indexed: 02/04/2023]
Abstract
Glutamate NMDA receptors (NMDARs) in the nucleus accumbens (NAc) are critically involved in drug dependence and reward. α2δ-1 is a newly discovered NMDAR-interacting protein that promotes synaptic trafficking of NMDARs independently of its conventional role as a calcium channel subunit. However, it remains unclear how repeated opioid exposure affects synaptic NMDAR activity and α2δ-1-NMDAR interaction in the NAc. In this study, whole-cell patch-clamp recordings showed that repeated treatment with morphine in mice markedly increased the NMDAR-mediated frequency of miniature excitatory postsynaptic currents (mEPSCs) and amplitude of puff NMDAR currents in medium spiny neurons in the NAc core region. Morphine treatment significantly increased the physical interaction of α2δ-1 with GluN1 and their synaptic trafficking in the NAc. In Cacna2d1 knockout mice, repeated treatment with morphine failed to increase the frequency of mEPSCs and amplitude of puff NMDAR currents in the NAc core. Furthermore, inhibition of α2δ-1 with gabapentin or disruption of the α2δ-1-NMDAR interaction with the α2δ-1 C terminus-interfering peptide blocked the morphine-elevated frequency of mEPSCs and amplitude of puff NMDAR currents in the NAc core. Correspondingly, systemically administered gabapentin, Cacna2d1 ablation, or microinjection of the α2δ-1 C terminus-interfering peptide into the NAc core attenuated morphine-induced conditioned place preference and locomotor sensitization. Our study reveals that repeated opioid exposure strengthens presynaptic and postsynaptic NMDAR activity in the NAc via α2δ-1. The α2δ-1-bound NMDARs in the NAc have a key function in the rewarding effect of opioids and could be targeted for treating opioid use disorder and addiction.
Collapse
Affiliation(s)
- Daozhong Jin
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hong Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shao-Rui Chen
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui-Lin Pan
- Center for Neuroscience and Pain Research, Department of Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
207
|
Viola TW, Orso R, Florian LF, Garcia MG, Gomes MGS, Mardini EM, Niederauer JPO, Zaparte A, Grassi‐Oliveira R. Effects of substance use disorder on oxidative and antioxidative stress markers: A systematic review and meta-analysis. Addict Biol 2023; 28:e13254. [PMID: 36577724 PMCID: PMC10078266 DOI: 10.1111/adb.13254] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/20/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
Recently, it has been suggested that central and peripheral toxicities identified in persons with substance use disorder (SUD) could be partially associated with an imbalance in reactive oxygen species and antioxidant defenses. We conducted a systematic review and meta-analysis to investigate whether SUD is associated with oxidative stress and to identify biomarkers possibly more affected by this condition. We have included studies that analysed oxidant and antioxidant markers in individuals with SUD caused by stimulants, alcohol, nicotine, opioids, and others (cannabis, inhalants, and polysubstance use). Our analysis showed that persons with SUD show higher oxidant markers and lower antioxidant markers than healthy controls. SUD was associated specifically with higher levels of oxidant markers malondialdehyde, thiobarbituric acid reactive substances and lipid peroxidation. Conversely, the antioxidant superoxide dismutase and the total antioxidant capacity/status were lowered in the SUD group. A meta-regression analysis revealed that persons with alcohol use disorder had higher oxidative stress estimates than those with stimulant use disorder. Moreover, individuals evaluated during abstinence showed smaller antioxidant effect sizes than non-abstinent ones. Our findings suggest a clear oxidative imbalance in persons with SUD, which could lead to cell damage and result in multiple associated comorbidities, particularly accelerated aging.
Collapse
Affiliation(s)
- Thiago Wendt Viola
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Rodrigo Orso
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Luisa Fossati Florian
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Miguel Gomes Garcia
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Marco Giovanni Signor Gomes
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Eduarda Mascarenhas Mardini
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - João Paulo Ottolia Niederauer
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
| | - Aline Zaparte
- LSU Pulmonary, Critical Care & Immunology, Department of MedicineLouisiana State University of Health SciencesNew OrleansLouisianaUSA
| | - Rodrigo Grassi‐Oliveira
- Developmental Cognitive Neuroscience Lab, School of Medicine, Brain Institute of the Rio Grande do Sul (BraIns)Pontifical Catholic University of Rio Grande do Sul (PUCRS)Porto AlegreBrazil
- Translational Neuropsychiatry Unit, Department of Clinical MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
208
|
Machado da Silva MC, Iglesias LP, Candelario-Jalil E, Khoshbouei H, Moreira FA, de Oliveira ACP. Role of Microglia in Psychostimulant Addiction. Curr Neuropharmacol 2023; 21:235-259. [PMID: 36503452 PMCID: PMC10190137 DOI: 10.2174/1570159x21666221208142151] [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: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022] Open
Abstract
The use of psychostimulant drugs can modify brain function by inducing changes in the reward system, mainly due to alterations in dopaminergic and glutamatergic transmissions in the mesocorticolimbic pathway. However, the etiopathogenesis of addiction is a much more complex process. Previous data have suggested that microglia and other immune cells are involved in events associated with neuroplasticity and memory, which are phenomena that also occur in addiction. Nevertheless, how dependent is the development of addiction on the activity of these cells? Although the mechanisms are not known, some pathways may be involved. Recent data have shown psychoactive substances may act directly on immune cells, alter their functions and induce various inflammatory mediators that modulate synaptic activity. These could, in turn, be involved in the pathological alterations that occur in substance use disorder. Here, we extensively review the studies demonstrating how cocaine and amphetamines modulate microglial number, morphology, and function. We also describe the effect of these substances in the production of inflammatory mediators and a possible involvement of some molecular signaling pathways, such as the toll-like receptor 4. Although the literature in this field is scarce, this review compiles the knowledge on the neuroimmune axis that is involved in the pathogenesis of addiction, and suggests some pharmacological targets for the development of pharmacotherapy.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Department of Pharmacology, Neuropharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil;
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Lia Parada Iglesias
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Habibeh Khoshbouei
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Fabrício Araujo Moreira
- Department of Pharmacology, Neuropsychopharmacology Laboratory, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | |
Collapse
|
209
|
Johnson BN, Kumar A, Su Y, Singh S, Sai KKS, Nader SH, Li S, Reboussin BA, Huang Y, Deep G, Nader MA. PET imaging of kappa opioid receptors and receptor expression quantified in neuron-derived extracellular vesicles in socially housed female and male cynomolgus macaques. Neuropsychopharmacology 2023; 48:410-417. [PMID: 36100655 PMCID: PMC9751296 DOI: 10.1038/s41386-022-01444-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/27/2022] [Accepted: 08/24/2022] [Indexed: 12/26/2022]
Abstract
Recent positron emission tomography (PET) studies of kappa opioid receptors (KOR) in humans reported significant relationships between KOR availability and social status, as well as cocaine choice. In monkey models, social status influences physiology, receptor pharmacology and behavior; these variables have been associated vulnerability to cocaine abuse. The present study utilized PET imaging to examine KOR availability in socially housed, cocaine-naïve female and male monkeys, and peripheral measures of KORs with neuron-derived extracellular vesicles (NDE). KOR availability was assessed in dominant and subordinate female and male cynomolgus macaques (N = 4/rank/sex), using PET imaging with the KOR selective agonist [11C]EKAP. In addition, NDE from the plasma of socially housed monkeys (N = 13/sex; N = 6-7/rank) were isolated by immunocapture method and analyzed for OPRK1 protein expression by ELISA. We found significant interactions between sex and social rank in KOR availability across 12 of 15 brain regions. This was driven by female data, in which KOR availability was significantly higher in subordinate monkeys compared with dominant monkeys; the opposite relationship was observed among males, but not statistically significant. No sex or rank differences were observed for NDE OPRK1 concentrations. In summary, the relationship between brain KOR availability and social rank was different in female and male monkeys. This was particularly true in female monkeys. We hypothesize that lower [11C]EKAP binding potentials were due to higher concentrations of circulating dynorphin, which is consistent with greater vulnerability in dominant compared with subordinate females. These findings suggest that the KOR is an important target for understanding the neurobiology associated with vulnerability to abused drugs and sex differences, and detectable in peripheral circulation.
Collapse
Affiliation(s)
- Bernard N Johnson
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ashish Kumar
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yixin Su
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Sangeeta Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Kiran Kumar Solingapuram Sai
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Susan H Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Songye Li
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Beth A Reboussin
- Department of Biostatistics and Data Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Gagan Deep
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | - Michael A Nader
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Center for Addiction Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
- Department of Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
210
|
Olasore HSA, Osuntoki AA, Magbagbeola OA, Awesu ARB, Olashore AA. Association of Dopamine Transporter Gene (DAT1) 40 bp 3′ UTR VNTR Polymorphism (rs28363170) and Cannabis Use Disorder. SUBSTANCE ABUSE: RESEARCH AND TREATMENT 2023; 17:11782218231163696. [PMID: 37020726 PMCID: PMC10068503 DOI: 10.1177/11782218231163696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 02/27/2023] [Indexed: 04/03/2023]
Abstract
Introduction: Cannabis remains the most widely used illicit drug among Nigerians, often associated with psychiatric disorders. Since genetic predisposition has been implicated in substance use disorders, we, therefore, aimed at finding out the relationship between dopamine transporter gene (DAT1) polymorphism and cannabis use disorder. Methods: We recruited 104 patients from a tertiary psychiatric facility in Lagos, Nigeria, who were diagnosed with cannabis use disorder according to ICD-10 and 96 non-smokers as a comparative group. The smokers were screened with Cannabis Use Disorder Identification Test (CUDIT), and cannabis dependence was assessed with the Severity of Dependence Scale (SDS). Genotyping was carried out for the 40 bp 3′ UTR VNTR of the DAT1 (rs28363170). Results: The frequencies of 9R/9R, 9R/10R, 10R/10R among non-smokers and smokers were 14 (14.3%), 25 (26.2%), 57 (59.5%) and 17 (16.3%), 54 (51.9%), 33 (31.7%) respectively. The genotype distribution was in Hardy Weinberg equilibrium (HWE) only in the smokers’ population (χ² = 1.896, P = .166). Individuals with the 10R allele were almost twice as likely as the 9R carriers to smoke cannabis (OR = 1.915, 95% CI: 1.225-2.995). However, this polymorphism was not associated with the quantity of cannabis smoked, age at onset of smoking, CUDIT, and SDS scores. Conclusion: The DAT VNTR polymorphism was associated with cannabis smoking but not cannabis use disorder.
Collapse
Affiliation(s)
- Holiness SA Olasore
- Department of Biochemistry, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Akinniyi A Osuntoki
- Department of Biochemistry, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Olubunmi A Magbagbeola
- Department of Biochemistry, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | | | - Anthony A Olashore
- Department of Psychiatry, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Psychiatry, University of Botswana, Gaborone, Botswana
| |
Collapse
|
211
|
Rendell MS. Obesity and diabetes: the final frontier. Expert Rev Endocrinol Metab 2023; 18:81-94. [PMID: 36710450 DOI: 10.1080/17446651.2023.2168643] [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: 06/28/2022] [Accepted: 01/11/2023] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Obesity is a key target in the treatment and prevention of diabetes and independently to reduce the burden of cardiovascular disease. We reviewed the options now available and anticipated to deal with obesity. AREAS COVERED We considered the epidemiology, genetics, and causation of obesity and the relationship to diabetes, and the dietary, pharmaceutical, and surgical management of the condition. The literature search covered both popular media via Google Search and the academic literature as indexed on PubMed with search terms including obesity, childhood obesity, adipocytes, insulin resistance, mechanisms of satiety, bariatric surgery, GLP-1 receptor agonists, and SGLT2 inhibitors. EXPERT OPINION Although bariatric surgery has been the primary approach to treating obese individuals, the emergence of agents impacting the brain satiety centers now promises effective, non-invasive treatment of obesity for individuals with and without diabetes. The GLP-1 receptor agonists have assumed the primary role in treating obesity with significant weight loss. Long-term results with semaglutide and tirzepatide are now approaching the success seen with bariatric surgery. Future agents combining the benefits of satiety control and thermogenesis to dissipate caloric excess are under investigation.
Collapse
Affiliation(s)
- Marc S Rendell
- The Association of Diabetes Investigators, Newport Coast, CA, USA
| |
Collapse
|
212
|
Bogdan R, Hatoum AS, Johnson EC, Agrawal A. The Genetically Informed Neurobiology of Addiction (GINA) model. Nat Rev Neurosci 2023; 24:40-57. [PMID: 36446900 PMCID: PMC10041646 DOI: 10.1038/s41583-022-00656-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
Abstract
Addictions are heritable and unfold dynamically across the lifespan. One prominent neurobiological theory proposes that substance-induced changes in neural circuitry promote the progression of addiction. Genome-wide association studies have begun to characterize the polygenic architecture undergirding addiction liability and revealed that genetic loci associated with risk can be divided into those associated with a general broad-spectrum liability to addiction and those associated with drug-specific addiction risk. In this Perspective, we integrate these genomic findings with our current understanding of the neurobiology of addiction to propose a new Genetically Informed Neurobiology of Addiction (GINA) model.
Collapse
Affiliation(s)
- Ryan Bogdan
- Department of Psychological and Brain Sciences, Washington University in St. Louis, St. Louis, MO, USA.
| | - Alexander S Hatoum
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Emma C Johnson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Arpana Agrawal
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
213
|
Poisel E, Zillich L, Streit F, Frank J, Friske MM, Foo JC, Mechawar N, Turecki G, Hansson AC, Nöthen MM, Rietschel M, Spanagel R, Witt SH. DNA methylation in cocaine use disorder-An epigenome-wide approach in the human prefrontal cortex. Front Psychiatry 2023; 14:1075250. [PMID: 36865068 PMCID: PMC9970996 DOI: 10.3389/fpsyt.2023.1075250] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/10/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Cocaine use disorder (CUD) is characterized by a loss of control over cocaine intake and is associated with structural, functional, and molecular alterations in the human brain. At the molecular level, epigenetic alterations are hypothesized to contribute to the higher-level functional and structural brain changes observed in CUD. Most evidence of cocaine-associated epigenetic changes comes from animal studies while only a few studies have been performed using human tissue. METHODS We investigated epigenome-wide DNA methylation (DNAm) signatures of CUD in human post-mortem brain tissue of Brodmann area 9 (BA9). A total of N = 42 BA9 brain samples were obtained from N = 21 individuals with CUD and N = 21 individuals without a CUD diagnosis. We performed an epigenome-wide association study (EWAS) and analyzed CUD-associated differentially methylated regions (DMRs). To assess the functional role of CUD-associated differential methylation, we performed Gene Ontology (GO) enrichment analyses and characterized co-methylation networks using a weighted correlation network analysis. We further investigated epigenetic age in CUD using epigenetic clocks for the assessment of biological age. RESULTS While no cytosine-phosphate-guanine (CpG) site was associated with CUD at epigenome-wide significance in BA9, we detected a total of 20 CUD-associated DMRs. After annotation of DMRs to genes, we identified Neuropeptide FF Receptor 2 (NPFFR2) and Kalirin RhoGEF Kinase (KALRN) for which a previous role in the behavioral response to cocaine in rodents is known. Three of the four identified CUD-associated co-methylation modules were functionally related to neurotransmission and neuroplasticity. Protein-protein interaction (PPI) networks derived from module hub genes revealed several addiction-related genes as highly connected nodes such as Calcium Voltage-Gated Channel Subunit Alpha1 C (CACNA1C), Nuclear Receptor Subfamily 3 Group C Member 1 (NR3C1), and Jun Proto-Oncogene, AP-1 Transcription Factor Subunit (JUN). In BA9, we observed a trend toward epigenetic age acceleration (EAA) in individuals with CUD remaining stable even after adjustment for covariates. CONCLUSION Results from our study highlight that CUD is associated with epigenome-wide differences in DNAm levels in BA9 particularly related to synaptic signaling and neuroplasticity. This supports findings from previous studies that report on the strong impact of cocaine on neurocircuits in the human prefrontal cortex (PFC). Further studies are needed to follow up on the role of epigenetic alterations in CUD focusing on the integration of epigenetic signatures with transcriptomic and proteomic data.
Collapse
Affiliation(s)
- Eric Poisel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lea Zillich
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Fabian Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Josef Frank
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marion M Friske
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jerome C Foo
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| | - Anita C Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stephanie H Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Center for Innovative Psychiatric and Psychotherapeutic Research, Biobank, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
214
|
Liu JL, Sun JT, Hu HL, Wang HY, Kang YX, Chen TQ, Chen ZH, Shang YX, Li YT, Hu B, Liu R. Structural and Functional Neural Alterations in Internet Addiction: A Study Protocol for Systematic Review and Meta-Analysis. Psychiatry Investig 2023; 20:69-74. [PMID: 36721888 PMCID: PMC9890045 DOI: 10.30773/pi.2021.0383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 07/04/2022] [Indexed: 01/26/2023] Open
Abstract
A growing number of neuroimaging studies have revealed abnormal brain structural and functional alterations in subjects with internet addiction (IA), however, with conflicting conclusions. We plan to conduct a systematic review and meta-analysis on the studies of voxelbased morphometry (VBM) and resting-state functional connectivity (rsFC), to reach a consolidated conclusion and point out the future direction in this field. A comprehensive search of rsFC and VBM studies of IA will be conducted in the PubMed, Cochrane Library, and Web of Science databases to retrieve studies published from the inception dates to August 2021. If the extracted data are feasible, activation likelihood estimation and seed-based d mapping methods will be used to meta-analyze the brain structural and functional changes in IA patients. This study will hopefully reach a consolidated conclusion on the impact of IA on human brain or point out the future direction in this field.
Collapse
Affiliation(s)
- Jun-Li Liu
- Xi'an Technological University, Xi'an, China
| | - Jing-Ting Sun
- Shaanxi University of Chinese Medicine, Xianyang, China.,Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui-Lin Hu
- Arizona State University, Tempe, AZ, USA
| | | | - Yun-Xi Kang
- Xi'an Technological University, Xi'an, China
| | - Tian-Qi Chen
- Institution of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Zhu-Hong Chen
- Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Xuan Shang
- Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu-Ting Li
- Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Bo Hu
- Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Rui Liu
- Department of Rehabilitation, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
215
|
Dieterich R, Endrass T. Neural Correlates of Cue Reactivity and the Regulation of Craving in Substance Use Disorders. ZEITSCHRIFT FUR KLINISCHE PSYCHOLOGIE UND PSYCHOTHERAPIE 2022. [DOI: 10.1026/1616-3443/a000680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract. Theoretical background: Considerable progress has been made in illuminating the neural basis of the compulsive use patterns characterizing substance use disorders. It has been suggested to utilize these findings to alleviate the health burden associated with substance use. Objective: We address how neuroimaging research can provide these benefits. Methods: Based on neurobiological models of addiction, we highlight neuroimaging research elucidating neural predictors of relapse and how treatments modify these markers. Results: With the focus on cue reactivity, brain activity related to the motivational salience of drugs and automatized use behaviors can predict relapse. Cue reactivity changes with abstinence, and it remains to be determined whether such changes confer periods of critical relapse susceptibility. Conclusions: Several established and emerging interventions modulate brain activity associated with drug value. However, executive deficits in addiction may compromise interventions targeting control-related prefrontal brain areas. Lastly, it remains more difficult to change the brain responses mediating habitual behaviors.
Collapse
Affiliation(s)
- Raoul Dieterich
- Addiction Research, Institute of Clinical Psychology and Psychotherapy, Faculty of Psychology, Technische Universität Dresden (TU Dresden), Germany
| | - Tanja Endrass
- Addiction Research, Institute of Clinical Psychology and Psychotherapy, Faculty of Psychology, Technische Universität Dresden (TU Dresden), Germany
| |
Collapse
|
216
|
Rosenthal A, Ebrahimi C, Wedemeyer F, Romanczuk-Seiferth N, Beck A. The Treatment of Substance Use Disorders: Recent Developments and New Perspectives. Neuropsychobiology 2022; 81:451-472. [PMID: 35724634 DOI: 10.1159/000525268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 04/28/2022] [Indexed: 11/19/2022]
Abstract
Substance-related disorders are complex psychiatric disorders that are characterized by continued consumption in spite of harmful consequences. Addiction affects various brain networks critically involved in learning, reward, and motivation, as well as inhibitory control. Currently applied therapeutic approaches aim at modification of behavior that ultimately leads to decrease of consumption or abstinence in individuals with substance use disorders. However, traditional treatment methods might benefit from recent neurobiological and cognitive neuroscientific research findings. Novel cognitive-behavioral approaches in the treatment of addictive behavior aim at enhancement of strategies to cope with stressful conditions as well as craving-inducing cues and target erroneous learning mechanisms, including cognitive bias modification, reconsolidation-based interventions, mindfulness-based interventions, virtual-reality-based cue exposure therapy as well as pharmacological augmentation strategies. This review discusses therapeutic strategies that target dysregulated neurocognitive processes associated with the development and maintenance of disordered substance use and may hold promise as effective treatments for substance-related disorders.
Collapse
Affiliation(s)
- Annika Rosenthal
- Department of Psychiatry and Neurosciences
- CCM, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Claudia Ebrahimi
- Department of Psychiatry and Neurosciences
- CCM, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Wedemeyer
- Department of Psychiatry and Neurosciences
- CCM, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nina Romanczuk-Seiferth
- Department of Psychiatry and Neurosciences
- CCM, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anne Beck
- Department of Psychiatry and Neurosciences
- CCM, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Health and Medical University, Campus Potsdam, Faculty of Health, Potsdam, Germany
| |
Collapse
|
217
|
Xu LX, Geng XM, Zhang JL, Guo XY, Potenza MN, Zhang JT. Neuromodulation treatments of problematic use of the Internet. Curr Opin Behav Sci 2022. [DOI: 10.1016/j.cobeha.2022.101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
218
|
Fang Y, Sun Y, Liu Y, Liu T, Hao W, Liao Y. Neurobiological mechanisms and related clinical treatment of addiction: a review. PSYCHORADIOLOGY 2022; 2:180-189. [PMID: 38665277 PMCID: PMC10917179 DOI: 10.1093/psyrad/kkac021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 04/28/2024]
Abstract
Drug addiction or substance use disorder (SUD), has been conceptualized as a three-stage (i.e. binge/intoxication, withdrawal/negative affect, and preoccupation/anticipation/craving) recurring cycle that involves complex changes in neuroplasticity, reward, motivation, desire, stress, memory, and cognitive control, and other related brain regions and brain circuits. Neuroimaging approaches, including magnetic resonance imaging, have been key to mapping neurobiological changes correlated to complex brain regions of SUD. In this review, we highlight the neurobiological mechanisms of these three stages of addiction. The abnormal activity of the ventral tegmental, nucleus accumbens, and caudate nucleus in the binge/intoxication stage involve the reward circuit of the midbrain limbic system. The changes in the orbitofrontal cortex, dorsolateral prefrontal cortex, amygdala, and hypothalamus emotional system in the withdrawal/negative affect stage involve increases in negative emotional states, dysphoric-like effects, and stress-like responses. The dysregulation of the insula and prefrontal lobes is associated with craving in the anticipation stage. Then, we review the present treatments of SUD based on these neuroimaging findings. Finally, we conclude that SUD is a chronically relapsing disorder with complex neurobiological mechanisms and multimodal stages, of which the craving stage with high relapse rate may be the key element in treatment efficacy of SUD. Precise interventions targeting different stages of SUD and characteristics of individuals might serve as a potential therapeutic strategy for SUD.
Collapse
Affiliation(s)
- Yehong Fang
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yunkai Sun
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yi Liu
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Tieqiao Liu
- Department of Psychiatry & Mental Health Institute of the Second Xiangya Hospital, Central South University. National Clinical Research Center on Mental Disorders & National Technology Institute on Mental Disorders. Hunan Key Laboratory of Psychiatry and Mental Health, 139 Renmin (M) Rd, Changsha, Hunan 410011, P. R. China
| | - Wei Hao
- Department of Psychiatry & Mental Health Institute of the Second Xiangya Hospital, Central South University. National Clinical Research Center on Mental Disorders & National Technology Institute on Mental Disorders. Hunan Key Laboratory of Psychiatry and Mental Health, 139 Renmin (M) Rd, Changsha, Hunan 410011, P. R. China
| | - Yanhui Liao
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| |
Collapse
|
219
|
Wang L, Wei L, Jin L, Li Y, Wei Y, He W, Shi L, Sun Q, Li W, Li Q, Li Y, Wu Y, Wang Y, Yuan M. Different Features of a Metabolic Connectivity Map and the Granger Causality Method in Revealing Directed Dopamine Pathways: A Study Based on Integrated PET/MR Imaging. AJNR Am J Neuroradiol 2022; 43:1770-1776. [PMID: 36357153 DOI: 10.3174/ajnr.a7707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/01/2022] [Indexed: 11/12/2022]
Abstract
BACKGROUND AND PURPOSE Exploring the directionality of neural information in the brain is important for understanding brain mechanisms and neurodisease development. Granger causality analysis and the metabolic connectivity map can be used to investigate directional transmission of information between brain regions, but their differences in depicting functional effective connectivity are not clear. MATERIALS AND METHODS Using the Monash rs-PET/MR imaging data set, we conducted Granger causality and metabolic connectivity map analyses of the dopamine reward circuit in the brain. The dopamine reward circuit is a well-known system consisting primarily of the bilateral orbital frontal cortex, caudate, nucleus accumbens, thalamus, and substantia nigra. We validated these circuit pathways using Granger causality and the metabolic connectivity map for identifying effective connectivities against a priori knowledge by testing the significance of directed pathways (P < .05, false discovery rate-corrected). RESULTS We found 3 types of effective connectivities in the dopamine reward circuit: long-range, neighborhood, and symmetric. Granger causality analysis revealed long-range connections in the orbital frontal cortex-caudate and orbital frontal cortex-nucleus accumbens regions. Metabolic connectivity map analysis revealed neighborhood connections in the nucleus accumbens-caudate, substantia nigra-thalamus, and thalamus-caudate regions. Metabolic connectivity map analysis also found symmetric connections in each of the bilateral nucleus accumbens, caudate, thalamus, and orbital frontal cortex-caudate regions. Different patterns in directional networks of the dopamine reward circuit were revealed by Granger causality and metabolic connectivity map analyses. CONCLUSIONS Granger causality analysis primarily identified bidirectional cortico-nucleus connections, while the metabolic connectivity map primarily identified direct connections among neighborhood and symmetric regions. The results of this study indicated that investigations of effective connectivities should use an appropriate analysis method depending on the purpose of the study.
Collapse
Affiliation(s)
- L Wang
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - L Wei
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - L Jin
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - Y Li
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - Y Wei
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - W He
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - L Shi
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| | - Q Sun
- Department of Radiology (Q.S., Y. Wang), the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - W Li
- Radiology (W.L., Q.L.), Tangdu Hospital of Air Force Military Medical University, Xi'an, China
| | - Q Li
- Radiology (W.L., Q.L.), Tangdu Hospital of Air Force Military Medical University, Xi'an, China
| | - Y Li
- Department of Radiology (YongBin Li), the First Hospital of Xi'an, Xi'an, China; and Siemens
| | - Y Wu
- Healthineers Ltd (Y. Wu), Beijing, China
| | - Y Wang
- Department of Radiology (Q.S., Y. Wang), the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - M Yuan
- From the Departments of Nuclear Medicine (L. Wang., L. Wei, L.J., YunBo Li, Y. Wei, W.H., L.S., M.Y.)
| |
Collapse
|
220
|
Hobkirk AL, Houser KR, Hoglen B, Bitzer ZT, Fendrich A, Bordner CR, Foulds J, Wang J, Mukherjee D, Yingst JM, Karunanayaka P, Goel R, Richie JP, Elias RJ, Yang QX. Evidence from an fMRI study that dessert-flavored e-cigarettes engage taste-related, but not smoking-related, brain circuitry for female daily smokers. Exp Clin Psychopharmacol 2022; 30:947-958. [PMID: 34110883 PMCID: PMC8660928 DOI: 10.1037/pha0000488] [Citation(s) in RCA: 2] [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: 12/24/2022]
Abstract
Regulations limiting the sale of flavored e-cigarette products are controversial for their potential to interfere with e-cigarette use as a cessation aid in addition to curbing youth use. Limited research suggests that flavor might enhance the addictive potential of e-cigarettes; however, the acute effects of flavored aerosols on brain function among humans have not been assessed. The present study aimed to isolate and compare the neural substrates of flavored and unflavored e-cigarette aerosols on brain function among nine female daily smokers. Participants inhaled aerosolized e-liquid with 36 mg/mL of nicotine with and without a strawberry-vanilla flavor while undergoing functional magnetic resonance imaging. We used general linear modeling to compare whole-brain mean neural activation and seed-to-voxel task-based functional connectivity between the flavored and unflavored inhalation runs. Contrary to our hypothesis, the flavored aerosol was associated with weaker activation than the unflavored aerosol in the brain stem and bilateral parietal-temporal-occipital region of the cortex. Instead, the flavor engaged taste-related brain regions while suppressing activation of the neural circuits typically engaged during smoking and nicotine administration. Alternatively, functional connectivity between subcortical dopaminergic brain seeds and cortical brain regions involved in motivation and reward salience were stronger during the flavored compared to unflavored aerosol run. The findings suggest that fruity and dessert-flavored e-cigarettes may dampen the reward experience of aerosol inhalation for smokers who initiate e-cigarette use by inhibiting activation of dopaminergic brain circuits. These preliminary findings may have implications for understanding how regulations on flavored e-cigarettes might impact their use as cessation aids. (PsycInfo Database Record (c) 2022 APA, all rights reserved).
Collapse
|
221
|
High Morphine Use Disorder Susceptibility Is Predicted by Impaired Learning Ability in Mice. Brain Sci 2022; 12:brainsci12121650. [PMID: 36552110 PMCID: PMC9776386 DOI: 10.3390/brainsci12121650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
An obvious reason for substance uses disorders (SUDs) is drug craving and seeking behavior induced by conditioned context, which is an abnormal solid context memory. The relationship between susceptibility to SUD and learning ability remains unclear in humans and animal models. In this study, we found that susceptibility to morphine use disorder (MUD) was negatively correlated with learning ability in conditioned place preference (CPP) in C57 mice. By using behavioral tests, we identified the FVB mouse as learning impaired. In addition, we discovered that learning-relevant proteins, such as the glutamate receptor subunits GluA1, NR1, and NR2A, were decreased in FVB mice. Finally, we assessed the context learning ability of FVB mice using the CPP test and priming. We found that FVB mice had lower learning performance with respect to normal memory but higher performance of morphine-reinstatement memory. Compared to C57 mice, FVB mice are highly sensitive to MUDs. Our results suggest that SUD susceptibility is predicted by impaired learning ability in mice; therefore, learning ability can play a simple and practical role in identifying high-risk SUD groups.
Collapse
|
222
|
Mowaad NA, El-Shamarka MEA, Khadrawy YA. The Behavioral and Neurochemical Changes Induced by Boldenone and/or Tramadol in Adult Male Rats. Neurochem Res 2022; 48:1320-1333. [PMID: 36449200 PMCID: PMC10066173 DOI: 10.1007/s11064-022-03827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/17/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022]
Abstract
AbstractBoldenone and tramadol are abused among large sectors of adolescents. Therefore, the behavioral changes concerned with memory and cognitive functions and neurochemical variations were investigated in the cortex of rats treated with boldenone and/or tramadol. Rats were divided into control and rats treated with boldenone, tramadol, or both drugs. At the end of the treatment period, the memory and cognitive functions were evaluated by the Y-maze test (YMT) and elevated plus maze test (EPMT) and the motor activity was determined by the open field test (OFT). The cortex was dissected to carry out the neurochemical analyses. Rats treated with boldenone and/or tramadol showed impaired memory and cognitive functions and reduced motor activity. A significant increase in lipid peroxidation (MDA), nitric oxide (NO), and a significant decrease in reduced glutathione (GSH) were observed in the cortex of rats treated with boldenone and/or tramadol. The levels of acetylcholinesterase (AChE) and monoamine oxidase (MAO) decreased significantly. Western blot data showed a significant decrease in Bcl2 and a significant increase in caspase-3 and inducible nitric oxide synthase (iNOS) in rats treated with boldenone and/or tramadol. These changes were associated with neuronal death as indicated from the histopathological examination.The present findings indicate that boldenone and/or tramadol induced impairment in memory and cognitive functions. These changes could be mediated by the increase in oxidative stress, neuroinflammation, reduced AChE level, and reduced number of survived neurons in the cortex as indicated from the decreased Bcl2 level and the histological examination.
Collapse
Affiliation(s)
- Noha A Mowaad
- Department of Narcotics, Ergogenic Aids and Poisons,Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marwa E A El-Shamarka
- Department of Narcotics, Ergogenic Aids and Poisons,Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Yasser A Khadrawy
- Medical Physiology Department, Medical Research and Clinical Studies Institute, National Research Centre, El-Behouth St, Giza, Egypt.
| |
Collapse
|
223
|
Li H, Hu T, Zhang Y, Zhao Z, Liu Q, Chen Z, Chen S. Extracellular signal-regulated kinase in the basolateral amygdala is required for reconsolidation of heroin-associated memory. Front Mol Neurosci 2022; 15:1020098. [PMID: 36438183 PMCID: PMC9684340 DOI: 10.3389/fnmol.2022.1020098] [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] [Received: 08/15/2022] [Accepted: 09/20/2022] [Indexed: 09/28/2023] Open
Abstract
Reconsolidation of heroin-associated memory is an independent memory process that occurs following retrieval, which is essential for the sustained capacity of an associative drug stimulus to precipitate heroin-seeking. Extracellular signal-regulated kinase (ERK) in the basolateral amygdala (BLA) mediates the reconsolidation of drug memory. In the present study, we utilized a rat model of drug craving and relapse to verify the hypothesis that the reconsolidation of heroin-associated memory requires ERK in an instrumental heroin-seeking behavior, focusing on the BLA brain region, which is crucial for synaptic plasticity and memory processes. We found that bilateral intra-BLA infusions of U0126 (1 μg/0.5 μl), an ERK inhibitor, immediately after retrieving heroin-associated memory significantly reduced cue-induced and drug-induced reinstatement and spontaneous recovery of heroin-seeking compared to the vehicle. Furthermore, this inhibitory effect was related to the characteristic of reconsolidation. Conversely, no effect was observed on the heroin-seeking behavior when the intra-BLA infusion of U0126 was administered 6 h after the heroin-associated memory retrieval or without memory retrieval. Together, these data suggest that disrupting the reconsolidation of heroin-associated memory via an ERK inhibitor may serve as a promising option for treating relapse in opiate addicts.
Collapse
Affiliation(s)
- Haoyu Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ting Hu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yanghui Zhang
- Center of Medical Genetics, Jiangmen Maternity and Child Health Care Hospital, Jiangmen, China
| | - Zijin Zhao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- The Institute of Skull Base Surgery and Neurooncology at Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zihua Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Si Chen
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Ophthalmology, Changsha, China
| |
Collapse
|
224
|
Effect of human mesenchymal stem cell secretome administration on morphine self-administration and relapse in two animal models of opioid dependence. Transl Psychiatry 2022; 12:462. [PMID: 36333316 PMCID: PMC9636200 DOI: 10.1038/s41398-022-02225-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
The present study investigates the possible therapeutic effects of human mesenchymal stem cell-derived secretome on morphine dependence and relapse. This was studied in a new model of chronic voluntary morphine intake in Wistar rats which shows classic signs of morphine intoxication and a severe naloxone-induced withdrawal syndrome. A single intranasal-systemic administration of MSCs secretome fully inhibited (>95%; p < 0.001) voluntary morphine intake and reduced the post-deprivation relapse intake by 50% (p < 0.02). Since several studies suggest a significant genetic contribution to the chronic use of many addictive drugs, the effect of MSCs secretome on morphine self-administration was further studied in rats bred as high alcohol consumers (UChB rats). Sub-chronic intraperitoneal administration of morphine before access to increasing concentrations of morphine solutions and water were available to the animals, led UChB rats to prefer ingesting morphine solutions over water, attaining levels of oral morphine intake in the range of those in the Wistar model. Intranasally administered MSCs secretome to UChB rats dose-dependently inhibited morphine self-administration by 72% (p < 0.001); while a single intranasal dose of MSC-secretome administered during a morphine deprivation period imposed on chronic morphine consumer UChB rats inhibited re-access morphine relapse intake by 80 to 85% (p < 0.0001). Both in the Wistar and the UChB rat models, MSCs-secretome administration reversed the morphine-induced increases in brain oxidative stress and neuroinflammation, considered as key engines perpetuating drug relapse. Overall, present preclinical studies suggest that products secreted by human mesenchymal stem cells may be of value in the treatment of opioid addiction.
Collapse
|
225
|
Migliaro M, Sánchez-Zavaleta R, Soto-Tinoco E, Ruiz-Contreras AE, Méndez-Díaz M, Herrera-Solís A, Pérez de la Mora M, Prospéro-García OE. Dominance status is associated with a variation in cannabinoid receptor 1 expression and amphetamine reward. Pharmacol Biochem Behav 2022; 221:173483. [PMID: 36270348 DOI: 10.1016/j.pbb.2022.173483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
The rewarding effects of psychostimulants appear to be distinct between dominant and subordinate individuals. In turn, the endocannabinoid system is an important modulator of drug reward in the nucleus accumbens and medial prefrontal cortex, however the connection with social dominance is yet to be established. Male rats were classified as dominant or subordinate on the basis of their spontaneous agonistic interactions and drug reward was assessed by means of conditioned place preference with amphetamine (AMPH). In addition, the expression of CB1R, CB2R, FAAH1, and DAGLa was quantified from accumbal and cortical tissue samples. Our findings demonstrate that dominant rats required a lesser dose of AMPH to acquire a preference for the drug-associated compartment, thereby suggesting a higher sensitivity to the rewarding effects of AMPH. Furthermore, dominants exhibited a lower expression of CB1R in the medial prefrontal cortex and nucleus accumbens. This study illustrates how CBR1 expression could differentiate the behavioral phenotypes associated to social dominance.
Collapse
Affiliation(s)
- Martin Migliaro
- Laboratorio de Cannabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Rodolfo Sánchez-Zavaleta
- Laboratorio de Cannabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Eva Soto-Tinoco
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Alejandra E Ruiz-Contreras
- Laboratorio de Neurogenómica Cognitiva, Coordinación de Psicobiología y Neurociencias, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Mónica Méndez-Díaz
- Laboratorio de Cannabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Andrea Herrera-Solís
- Laboratorio de Efectos Terapéuticos de los Cannabinoides, Subdirección de Investigación Biomédica, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico
| | - Miguel Pérez de la Mora
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Oscar E Prospéro-García
- Laboratorio de Cannabinoides, Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
226
|
Hill-Bowen LD, Riedel MC, Salo T, Flannery JS, Poudel R, Laird AR, Sutherland MT. Convergent gray matter alterations across drugs of abuse and network-level implications: A meta-analysis of structural MRI studies. Drug Alcohol Depend 2022; 240:109625. [PMID: 36115222 DOI: 10.1016/j.drugalcdep.2022.109625] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/01/2022] [Accepted: 09/06/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Neuroimaging studies often consider brain alterations linked with substance abuse within the context of individual drugs (e.g., nicotine), while neurobiological theories of addiction emphasize common brain network-level alterations across drug classes. Using emergent meta-analytic techniques, we identified common structural brain alterations across drugs and characterized the functionally-connected networks with which such structurally altered regions interact. METHODS We identified 82 articles characterizing gray matter (GM) volume differences for substance users vs. controls. Using the anatomical likelihood estimation algorithm, we identified convergent GM reductions across drug classes. Next, we performed resting-state and meta-analytic functional connectivity analyses using each structurally altered region as a seed and computed whole-brain functional connectivity profiles as the union of both maps. We characterized an "extended network" by identifying brain areas demonstrating the highest degree of functional coupling with structurally impacted regions. Finally, hierarchical clustering was performed leveraging extended network nodes' functional connectivity profiles to delineate subnetworks. RESULTS Across drug classes, we identified medial frontal/ventromedial prefrontal, and multiple regions in anterior cingulate (ACC) and insula as regions displaying convergent GM reductions among users. Overlap of these regions' functional connectivity profiles identified ACC, inferior frontal, PCC, insula, superior temporal, and putamen as regions of an impacted extended network. Hierarchical clustering revealed 3 subnetworks closely corresponding to default mode (PCC, angular), salience (dACC, caudate), and executive control networks (dlPFC and parietal). CONCLUSIONS These outcomes suggest that substance-related structural brain alterations likely have implications for the functioning of canonical large-scale networks and the perpetuation of substance use and neurocognitive alterations.
Collapse
Affiliation(s)
- Lauren D Hill-Bowen
- Department of Psychology, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States
| | - Michael C Riedel
- Department of Physics, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States
| | - Taylor Salo
- Department of Psychology, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States
| | - Jessica S Flannery
- Department of Psychology and Neuroscience, University of North Carolina, Chapel Hill, NC, United States
| | - Ranjita Poudel
- Department of Psychology, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States
| | - Angela R Laird
- Department of Physics, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States
| | - Matthew T Sutherland
- Department of Psychology, Florida International University, 11200 SW 8th Street, Miami, FL 33199, United States.
| |
Collapse
|
227
|
Petit B, Dornier A, Meille V, Demina A, Trojak B. Non-invasive brain stimulation for smoking cessation: a systematic review and meta-analysis. Addiction 2022; 117:2768-2779. [PMID: 35470522 DOI: 10.1111/add.15889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 03/15/2022] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIMS Non-invasive brain stimulation (NIBS) methods have showed promising results for the treatment of tobacco use disorder, but little is known about the efficacy of NIBS on sustained tobacco abstinence. We aimed to assess its effectiveness for long-term smoking cessation. METHODS Systematic review and meta-analysis of randomized controlled trials (RCT). PubMed, Cochrane library, Embase, PsycINFO and clinical trials registries were systematically searched for relevant studies up to May 2021. Relevant studies included adult smokers seeking smoking cessation, included in an RCT using NIBS [specifically repetitive transcranial magnetic stimulation (rTMS) and transcranial direct current stimulation (tDCS)], and with follow-up of more than 4 weeks. There were no restrictions on location. Abstinence rates in the active NIBS groups were compared with abstinence rates in sham NIBS or in usual treatment groups, from 4 weeks to 12 months following the quit attempt. Smoking abstinence was measured on an intention-to-treat basis and we used risk ratios (RRs) as measures of effect size. RESULTS Seven studies were included (n = 699 patients). In all included studies, the control groups were receiving sham NIBS and only data from 3 to 6 months were analysable. By pooling the seven included studies, the RR of sustained abstinence of any form of NIBS relative to sham NIBS was 2.39 [95% confidence interval (CI) = 1.26-4.55; I2 = 40%]. Subgroup analyses found that the RR was even higher when excitatory rTMS was used on the left dorsolateral prefrontal cortex (RR = 4.34; 95% CI = 1.69-11.18; I2 = 0%) or when using deep rTMS targeting the lateral prefrontal cortex and insula bilaterally (RR = 4.64; 95% CI = 1.61-13.39; I2 = 0%). A high risk of bias was found in four included studies. We also determined, using grades of recommendation, assessment, development and evaluation, that overall there was a low level of confidence in the results. CONCLUSION Non-invasive brain stimulation (NIBS) may improve smoking abstinence rates from 3 to 6 months after quitting smoking, compared with sham NIBS or usual treatment.
Collapse
Affiliation(s)
- Benjamin Petit
- Department of Addictology, University Hospital of Dijon, Dijon, France
| | - Alexandre Dornier
- Department of Addictology, University Hospital of Dijon, Dijon, France
| | - Vincent Meille
- Department of Addictology, University Hospital of Dijon, Dijon, France
| | - Anastasia Demina
- Department of Addictology, University Hospital of Dijon, Dijon, France
| | - Benoit Trojak
- Department of Addictology, University Hospital of Dijon, Dijon, France.,University of Burgundy, Cognition, Action et Plasticité Sensorimotrice, Dijon, France
| |
Collapse
|
228
|
Liu QQ, Cheng YX, Jing Q, Zhang KM, Ding LF, Fan XW, Jia CH, Xu F, Bi GQ, Lau PM. Preferential subcortical collateral projections of pedunculopontine nucleus-targeting cortical pyramidal neurons revealed by brain-wide single fiber tracing. Mol Brain 2022; 15:88. [PMID: 36309684 PMCID: PMC9618196 DOI: 10.1186/s13041-022-00975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/24/2022] [Indexed: 12/29/2022] Open
Abstract
The pedunculopontine nucleus (PPN) is a heterogeneous midbrain structure involved in various brain functions, such as motor control, learning, reward, and sleep. Previous studies using conventional tracers have shown that the PPN receives extensive afferent inputs from various cortical areas. To examine how these cortical axons make collateral projections to other subcortical areas, we used a dual-viral injection strategy to sparsely label PPN-targeting cortical pyramidal neurons in CaMKIIα-Cre transgenic mice. Using a high-speed volumetric imaging with on-the-fly-scan and Readout (VISoR) technique, we visualized brain-wide axonal projections of individual PPN-targeting neurons from several cortical areas, including the prelimbic region (PL), anterior cingulate area (ACA) and secondary motor cortex (MOs). We found that each PPN-projecting neuron had a unique profile of collateralization, with some subcortical areas being preferential targets. In particular, PPN-projecting neurons from all three traced cortical areas exhibited common preferential collateralization to several nuclei, with most neurons targeting the striatum (STR), lateral hypothalamic area (LHA) and periaqueductal gray (PAG), and a substantial portion of neurons also targeting the zona incerta (ZI), median raphe nucleus (MRN) and substantia nigra pars reticulata (SNr). Meanwhile, very specific collateralization patterns were found for other nuclei, including the intermediate reticular nucleus (IRN), parvicellular reticular nucleus (PARN) and gigantocellular reticular nucleus (GRN), which receive collateral inputs almost exclusively from the MOs. These observations provide potential anatomical mechanisms for cortical neurons to coordinate the PPN with other subcortical areas in performing different physiological functions.
Collapse
Affiliation(s)
- Qiao-Qiong Liu
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Yu-Xiao Cheng
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Qi Jing
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Ke-Ming Zhang
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Lu-Feng Ding
- grid.458489.c0000 0001 0483 7922Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| | - Xiao-Wei Fan
- grid.458489.c0000 0001 0483 7922Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| | - Chun-Hui Jia
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China
| | - Fang Xu
- grid.458489.c0000 0001 0483 7922Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| | - Guo-Qiang Bi
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China ,grid.458489.c0000 0001 0483 7922Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| | - Pak-Ming Lau
- grid.59053.3a0000000121679639Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026 Anhui China ,grid.458489.c0000 0001 0483 7922Interdisciplinary Center for Brain Information, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 Guangdong China
| |
Collapse
|
229
|
Vamvakopoulou IA, Fonville L, Hayes A, McGonigle J, Elliott R, Ersche KD, Flechais R, Orban C, Murphy A, Smith DG, Suckling J, Taylor EM, Deakin B, Robbins TW, Nutt DJ, Lingford-Hughes AR, Paterson LM. Selective D3 receptor antagonism modulates neural response during negative emotional processing in substance dependence. Front Psychiatry 2022; 13:998844. [PMID: 36339857 PMCID: PMC9627287 DOI: 10.3389/fpsyt.2022.998844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Negative affective states contribute to the chronic-relapsing nature of addiction. Mesolimbic dopamine D3 receptors are well placed to modulate emotion and are dysregulated in substance dependence. Selective antagonists might restore dopaminergic hypofunction, thus representing a potential treatment target. We investigated the effects of selective D3 antagonist, GSK598809, on the neural response to negative emotional processing in substance dependent individuals and healthy controls. Methodology Functional MRI BOLD response was assessed during an evocative image task, 2 h following acute administration of GSK598809 (60 mg) or placebo in a multi-site, double-blind, pseudo-randomised, cross-over design. Abstinent drug dependent individuals (DD, n = 36) comprising alcohol-only (AO, n = 19) and cocaine-alcohol polydrug (PD, n = 17) groups, and matched controls (n = 32) were presented with aversive and neutral images in a block design (contrast of interest: aversive > neutral). Whole-brain mixed-effects and a priori ROI analyses tested for group and drug effects, with identical models exploring subgroup effects. Results No group differences in task-related BOLD signal were identified between DD and controls. However, subgroup analysis revealed greater amygdala/insular BOLD signal in PD compared with AO groups. Following drug administration, GSK598809 increased BOLD response across HC and DD groups in thalamus, caudate, putamen, and pallidum, and reduced BOLD response in insular and opercular cortices relative to placebo. Multivariate analyses in a priori ROIs revealed differential effects of D3 antagonism according to subgroup in substantia nigra; GSK598809 increased BOLD response in AO and decreased response in PD groups. Conclusion Acute GSK598809 modulates the BOLD response to aversive image processing, providing evidence that D3 antagonism may impact emotional regulation. Enhanced BOLD response within D3-rich mesolimbic regions is consistent with its pharmacology and with attenuation of substance-related hypodopaminergic function. However, the lack of group differences in task-related BOLD response and the non-specific effect of GSK598809 between groups makes it difficult to ascertain whether D3 antagonism is likely to be normalising or restorative in our abstinent populations. The suggestion of differential D3 modulation between AO and PD subgroups is intriguing, raising the possibility of divergent treatment responses. Further study is needed to determine whether D3 antagonism should be recommended as a treatment target in substance dependence.
Collapse
Affiliation(s)
- Ioanna A. Vamvakopoulou
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Leon Fonville
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Alexandra Hayes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - John McGonigle
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Rebecca Elliott
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Karen D. Ersche
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Remy Flechais
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Csaba Orban
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Anna Murphy
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Dana G. Smith
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - John Suckling
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor M. Taylor
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Bill Deakin
- Neuroscience and Psychiatry Unit, Institute of Brain, Behaviour and Mental Health, The University of Manchester, Manchester, United Kingdom
| | - Trevor W. Robbins
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| | - David J. Nutt
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Anne R. Lingford-Hughes
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Louise M. Paterson
- Division of Psychiatry, Department of Brain Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
230
|
Addictive and other mental disorders: a call for a standardized definition of dual disorders. Transl Psychiatry 2022; 12:446. [PMID: 36229453 PMCID: PMC9562408 DOI: 10.1038/s41398-022-02212-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
The persistent difficulty in conceptualizing the relationship between addictive and other mental disorders stands out among the many challenges faced by the field of Psychiatry. The different philosophies and schools of thought about, and the sheer complexity of these highly prevalent clinical conditions make progress inherently difficult, not to mention the profusion of competing and sometimes contradictory terms that unnecessarily exacerbate the challenge. The lack of a standardized term adds confusion, fuels stigma, and contributes to a "wrong door syndrome" that captures the difficulty of not only diagnosing but also treating addictive and other mental disorders in an integrated manner. The World Association on Dual Disorders (WADD) proposes the adoption of the term "Dual Disorder" which, while still arbitrary, would help harmonize various clinical and research efforts by rallying around a single, more accurate, and less stigmatizing designation.
Collapse
|
231
|
Meyer D, Athota P, Gowen A, Nguyen NM, Schaal VL, Yelamanchili SV, Pendyala G. Effect of Combined Methamphetamine and Oxycodone Use on the Synaptic Proteome in an In Vitro Model of Polysubstance Use. Genes (Basel) 2022; 13:genes13101816. [PMID: 36292701 PMCID: PMC9601452 DOI: 10.3390/genes13101816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 01/03/2023] Open
Abstract
Polysubstance use (PSU) generally involves the simultaneous use of an opioid along with a stimulant. In recent years, this problem has escalated into a nationwide epidemic. Understanding the mechanisms and effects underlying the interaction between these drugs is essential for the development of treatments for those suffering from addiction. Currently, the effect of PSU on synapses-critical points of contact between neurons-remains poorly understood. Using an in vitro model of primary neurons, we examined the combined effects of the psychostimulant methamphetamine (METH) and the prescription opioid oxycodone (oxy) on the synaptic proteome using quantitative mass-spectrometry-based proteomics. A further ClueGO analysis and Ingenuity Pathway Analysis (IPA) indicated the dysregulation of several molecular functions, biological processes, and pathways associated with neural plasticity and structural development. We identified one key synaptic protein, Striatin-1, which plays a vital role in many of these processes and functions, to be downregulated following METH+oxy treatment. This downregulation of Striatin-1 was further validated by Western blot. Overall, the present study indicates several damaging effects of the combined use of METH and oxy on neural function and warrants further detailed investigation into mechanisms contributing to synaptic dysfunction.
Collapse
Affiliation(s)
- Daniel Meyer
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pranavi Athota
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Austin Gowen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nghi M. Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Victoria L. Schaal
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sowmya V. Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
- National Strategic Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-8690
| |
Collapse
|
232
|
Ruedisueli I, Arastoo S, Gupta PK, Gornbein J, Middlekauff H. Neural-hematopoietic-inflammatory axis in nonsmokers, electronic cigarette users, and tobacco smokers. Physiol Rep 2022; 10:e15412. [PMID: 36200129 PMCID: PMC9535258 DOI: 10.14814/phy2.15412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 06/29/2022] [Accepted: 07/14/2022] [Indexed: 06/16/2023] Open
Abstract
Amygdala activity in context of the splenocardiac model has not been investigated in healthy, young adults and has not been compared between nonsmokers, electronic cigarette users, and smokers. The purpose of the current study was to determine whether fluorodeoxyglucose positron emission tomography/computer tomography (FDG PET/CT) scans would demonstrate positively correlated metabolic activity in the amygdala, bone marrow, spleen, and aorta, elucidating activation of the splenocardiac axis in otherwise healthy young people who use tobacco products compared to nonusers. Moreover, the study was conducted to evaluate whether electronic cigarette users and tobacco smokers have similar levels of inflammation compared to nonusers. In 45 healthy adults (mean age = 25 years), including nonsmoker (n = 15), electronic cigarette user (n = 16), and smoker (n = 14) groups, metabolic activity in the amygdala, spleen, aorta, bone marrow of thoracic vertebrae, and adjacent erector spinae skeletal muscle was quantified through visualization of radioactive glucose (18 FDG) uptake by FDG-PET/CT. The maximum standardized uptake value for each region was calculated for correlation analyses and comparisons between groups. In correlation analyses, metabolic activity of the amygdala correlated with metabolic activity in the aorta (r = 0.757), bone marrow (r = 0.750), and spleen (r = 0.665), respectively. Metabolic activity in the aorta correlated with 18 FDG uptake in the thoracic vertebrae (r = 0.703) and spleen (r = 0.594), respectively. Metabolic activity in the spleen also correlated with 18 FDG uptake in the bone marrow (r = 0.620). Metabolic activity in the adjacent erector spinae skeletal muscle (our control tissue) was not positively correlated with any other region of interest. Finally, there were no statistically significant mean differences in metabolic activity between the three groups: nonsmokers, electronic cigarette users, and smokers in any target tissue. Amygdala metabolic activity, as measured by 18 FDG uptake in FDG-PET/CT scans, positively correlated with inflammation in the splenocardiac tissues, including: the aorta, bone marrow, and spleen, underscoring the existence of a neural-hematopoietic-inflammatory axis in healthy, young adults.
Collapse
Affiliation(s)
- Isabelle Ruedisueli
- Division of Cardiology, Department of MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Sara Arastoo
- Department of Anesthesiology and Perioperative MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Pawan K. Gupta
- Department of Molecular and Medical PharmacologyDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Jeffrey Gornbein
- Departments of Medicine and Computational MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| | - Holly R. Middlekauff
- Division of Cardiology, Department of MedicineDavid Geffen School of Medicine at UCLALos AngelesCaliforniaUSA
| |
Collapse
|
233
|
Ghasemian S, Vardanjani MM, Sheibani V, Mansouri FA. Dynamic modulation of inhibition ability following repeated exposures to morphine in macaque monkey. J Psychopharmacol 2022; 36:1151-1160. [PMID: 35971887 DOI: 10.1177/02698811221112952] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Deficits in cognitive control, particularly inhibition ability, play crucial roles in susceptibility, progress, and relapse to opioid addiction. However, it is unclear when and how such deficits develop and interact with repeated exposures to prescribed opioids. AIM Using macaque monkey (Macaca mulatta), as an animal model with high translational merits in cognitive neuroscience, we tried to delineate alterations of inhibition ability in the course of repeated exposures to morphine. METHODS Monkeys were trained to perform stop-signal task and then we closely monitored their inhibition ability before exposure, after initial exposure, and following repeated exposures to morphine when they experienced abstinent periods. We also assessed morphine-induced conditioned place preference (CPP) in these monkeys to monitor the long-lasting effects of morphine on other behaviors. RESULTS Compared to the baseline level, monkeys' inhibition ability was significantly enhanced after initial exposure to morphine (early phase); however, it became significantly attenuated after repeated exposures (late phase). These alterations occurred while monkeys consistently expressed the morphine-induced CPP over the course of morphine exposure. CONCLUSIONS Our findings indicate that repeated and scheduled exposures to morphine, which is akin to its clinical and recreational use, lead to dynamic alterations in primates' cognitive control depending on the extent of exposure. Enhancement of inhibition after limited exposure might provide opportunities to intervene and prevent the progress and culmination of opioid addiction, which is characterized by disinhibited drug-seeking and consumption.
Collapse
Affiliation(s)
- Sadegh Ghasemian
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Cognitive Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Marzieh M Vardanjani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Cognitive Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Cognitive Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farshad A Mansouri
- ARC Centre of Excellence for Integrative Brain function, Monash University, Clayton, VIC, Australia
| |
Collapse
|
234
|
Brandt L, Liu S, Heim C, Heinz A. The effects of social isolation stress and discrimination on mental health. Transl Psychiatry 2022; 12:398. [PMID: 36130935 PMCID: PMC9490697 DOI: 10.1038/s41398-022-02178-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Social isolation and discrimination are growing public health concerns associated with poor physical and mental health. They are risk factors for increased morbidity and mortality and reduced quality of life. Despite their detrimental effects on health, there is a lack of knowledge regarding translation across the domains of experimental research, clinical studies, and real-life applications. Here, we review and synthesize evidence from basic research in animals and humans to clinical translation and interventions. Animal models indicate that social separation stress, particularly in early life, activates the hypothalamic-pituitary-adrenal axis and interacts with monoaminergic, glutamatergic, and GABAergic neurotransmitter systems, inducing long-lasting reductions in serotonin turnover and alterations in dopamine receptor sensitivity. These findings are of particular importance for human social isolation stress, as effects of social isolation stress on the same neurotransmitter systems have been implicated in addictive, psychotic, and affective disorders. Children may be particularly vulnerable due to lasting effects of social isolation and discrimination stress on the developing brain. The effects of social isolation and loneliness are pronounced in the context of social exclusion due to discrimination and racism, during widespread infectious disease related containment strategies such as quarantine, and in older persons due to sociodemographic changes. This highlights the importance of new strategies for social inclusion and outreach, including gender, culture, and socially sensitive telemedicine and digital interventions for mental health care.
Collapse
Affiliation(s)
- Lasse Brandt
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin (Campus Charité Mitte), Berlin, Germany
| | - Shuyan Liu
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin (Campus Charité Mitte), Berlin, Germany
| | - Christine Heim
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin (Campus Charité Mitte), Berlin, Germany
| | - Andreas Heinz
- Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin (Campus Charité Mitte), Berlin, Germany.
| |
Collapse
|
235
|
Imaging the Limbic System in Parkinson's Disease-A Review of Limbic Pathology and Clinical Symptoms. Brain Sci 2022; 12:brainsci12091248. [PMID: 36138984 PMCID: PMC9496800 DOI: 10.3390/brainsci12091248] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/05/2022] [Accepted: 09/13/2022] [Indexed: 01/09/2023] Open
Abstract
The limbic system describes a complex of brain structures central for memory, learning, as well as goal directed and emotional behavior. In addition to pathological studies, recent findings using in vivo structural and functional imaging of the brain pinpoint the vulnerability of limbic structures to neurodegeneration in Parkinson's disease (PD) throughout the disease course. Accordingly, dysfunction of the limbic system is critically related to the symptom complex which characterizes PD, including neuropsychiatric, vegetative, and motor symptoms, and their heterogeneity in patients with PD. The aim of this systematic review was to put the spotlight on neuroimaging of the limbic system in PD and to give an overview of the most important structures affected by the disease, their function, disease related alterations, and corresponding clinical manifestations. PubMed was searched in order to identify the most recent studies that investigate the limbic system in PD with the help of neuroimaging methods. First, PD related neuropathological changes and corresponding clinical symptoms of each limbic system region are reviewed, and, finally, a network integration of the limbic system within the complex of PD pathology is discussed.
Collapse
|
236
|
Al-Moussally F, Fogel EL, Helft PR. An ethical analysis of endoscopic therapy decision-making in patients with refractory substance use disorder and chronic pancreatitis. Pancreatology 2022; 22:671-677. [PMID: 35691886 PMCID: PMC10118247 DOI: 10.1016/j.pan.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 05/15/2022] [Accepted: 05/30/2022] [Indexed: 12/11/2022]
Affiliation(s)
- Feras Al-Moussally
- Indiana University School of Medicine, United States; Charles Warren Fairbanks Center of Medical Ethics, Indiana University Health, United States
| | - Evan L Fogel
- Indiana University School of Medicine, United States; Lehman, Bucksot and Sherman Section of Pancreatobiliary Endoscopy, United States
| | - Paul R Helft
- Indiana University School of Medicine, United States; Charles Warren Fairbanks Center of Medical Ethics, Indiana University Health, United States.
| |
Collapse
|
237
|
Greener MR, Storr SJ. Exploring the Role of DARPP-32 in Addiction: A Review of the Current Limitations of Addiction Treatment Pathways and the Role of DARPP-32 to Improve Them. NEUROSCI 2022; 3:494-509. [PMID: 39483434 PMCID: PMC11523713 DOI: 10.3390/neurosci3030035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2024] Open
Abstract
We are amidst a global addiction crisis, yet stigmas surrounding addiction counterintuitively prevail. Understanding and appreciating the neurobiology of addiction is essential to dissolve this stigma and for the development of new pharmacological agents to improve upon currently narrow therapeutic options. This review highlights this and evaluates dopamine-and-cAMP-regulated phosphoprotein, Mr 32 kDa (DARPP-32) as a potential target to treat various forms of substance abuse. Despite the proven involvement of DARPP-32 in addiction pathophysiology, no robust investigations into compounds that could pharmacologically modulate it have been carried out. Agents capable of altering DARPP-32 signalling in this way could prevent or reverse drug abuse and improve upon currently substandard treatment options.
Collapse
Affiliation(s)
- Megan R. Greener
- Biodiscovery Institute Phase 3, Entrance 2, Building 43, University of Nottingham, University Park, Nottingham NG7 2RD, UK
| | | |
Collapse
|
238
|
Design, Synthesis and Pharmacological Evaluation of Novel Conformationally Restricted N-arylpiperazine Derivatives Characterized as D 2/D 3 Receptor Ligands, Candidates for the Treatment of Neurodegenerative Diseases. Biomolecules 2022; 12:biom12081112. [PMID: 36009006 PMCID: PMC9405847 DOI: 10.3390/biom12081112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Most neurodegenerative diseases are multifactorial, and the discovery of several molecular mechanisms related to their pathogenesis is constantly advancing. Dopamine and dopaminergic receptor subtypes are involved in the pathophysiology of several neurological disorders, such as schizophrenia, depression and drug addiction. For this reason, the dopaminergic system and dopamine receptor ligands play a key role in the treatment of such disorders. In this context, a novel series of conformationally restricted N-arylpiperazine derivatives (5a–f) with a good affinity for D2/D3 dopamine receptors is reported herein. Compounds were designed as interphenylene analogs of the drugs aripiprazole (2) and cariprazine (3), presenting a 1,3-benzodioxolyl subunit as a ligand of the secondary binding site of these receptors. The six new N-arylpiperazine compounds were synthesized in good yields by using classical methodologies, and binding and guanosine triphosphate (GTP)-shift studies were performed. Affinity values below 1 μM for both target receptors and distinct profiles of intrinsic efficacy were found. Docking studies revealed that Compounds 5a–f present a different binding mode with dopamine D2 and D3 receptors, mainly as a consequence of the conformational restriction imposed on the flexible spacer groups of 2 and 3.
Collapse
|
239
|
Psychiatric Comorbidities Associated with Persistent Postoperative Opioid Use. Curr Pain Headache Rep 2022; 26:701-708. [PMID: 35960447 DOI: 10.1007/s11916-022-01073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW This review outlines the psychiatric comorbidities associated with persistent opioid use in the postoperative period. We finish our analysis with evidence-based, patient-centered interventions that can be rendered in the perioperative setting to decrease postoperative opioid requirements. RECENT FINDINGS Opioids are overprescribed in the USA, especially in the postoperative setting. Excess opioids can result in diversion and contribute to the ongoing opioid epidemic. Mental health and substance use disorders can contribute to persistent postoperative opioid use. Adequately managing these disorders preoperatively promises to reduce persistent postoperative opioid use. Due to the lack of homogenous, evidence-based recommendations on the appropriate quantity and duration of postoperative opioid therapy, there is wide variability in provider prescribing habits. Further research is needed to establish surgery-specific postoperative opioid therapy protocols. Opioids continue to be a mainstay in the treatment of postoperative pain. Unmonitored postoperative opioid use can lead to opioid use disorder. Mental health disorders increase susceptibility to persistent postoperative opioid use. By managing these psychiatric illnesses preoperatively, clinicians have the ability to decrease opioid consumption postoperatively. Lastly, given the healthcare burden of opioid misuse and abuse, it is important to establish concrete protocols to guide provider-prescribing habits.
Collapse
|
240
|
Lu L, Yang W, Zhang X, Tang F, Du Y, Fan L, Luo J, Yan C, Zhang J, Li J, Liu J, von Deneen KM, Yu D, Liu J, Yuan K. Potential brain recovery of frontostriatal circuits in heroin users after prolonged abstinence: A preliminary study. J Psychiatr Res 2022; 152:326-334. [PMID: 35785575 DOI: 10.1016/j.jpsychires.2022.06.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022]
Abstract
Neuroscientists have devoted efforts to explore potential brain recovery after prolonged abstinence in heroin users (HU). However, not much is known about whether frontostriatal circuits can recover after prolonged abstinence in HU. An eight-month longitudinal study was carried out for HU. Two MRI scans were obtained at baseline (HU1) and 8-month follow-up (HU2). The functional and structural connectivities of dorsal and ventral frontostriatal pathways were measured by resting-state functional connectivity (RSFC) and diffusion tensor imaging (DTI). Correlation analyses were employed to reveal the associations between neuroimaging and behavioral changes. Results suggested that relative to healthy controls (HCs), HU1 showed lower fractional anisotropy (FA) in the right dorsolateral prefrontal cortex (DLPFC)-to-caudate tracts and medial orbitofrontal cortex (mOFC)-to-nucleus accumbens (NAc) tracts as well as decreased RSFC in the left mOFC-NAc circuits. Longitudinal results revealed reduced craving and enhanced cognitive control in HU2 compared with HU1. After prolonged abstinence, HU2 showed increased FA values in the right DLPFC-caudate and mOFC-NAc tracts as well as increased RSFC strength in the bilateral mOFC-NAc circuits compared with HU1. In addition, changes in RSFC and FA values in the right mOFC-NAc circuit were negatively correlated with craving score changes. Similarly, negative correlations were also found between changes of RSFC in the bilateral DLPFC-caudate circuits and TMT-A scores. We provided scientific evidence for brain recovery of the dorsal and ventral frontostriatal circuits in HU after prolonged abstinence, and these circuits may be potential neuroimaging biomarkers for cognition and craving changes.
Collapse
Affiliation(s)
- Ling Lu
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Wenhan Yang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Xiaozi Zhang
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Fei Tang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Yanyao Du
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Fan
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Jing Luo
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Cui Yan
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China
| | - Jun Zhang
- Hunan Judicial Police Academy, Changsha, China
| | - Jun Li
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Jixin Liu
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi, 710071, China
| | - Karen M von Deneen
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Dahua Yu
- International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, China.
| | - Kai Yuan
- Center for Brain Imaging, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, Xi'an, Shaanxi, 710071, China; International Joint Research Center for Advanced Medical Imaging and Intelligent Diagnosis and Treatment & Xi'an Key Laboratory of Intelligent Sensing and Regulation of trans-Scale Life Information, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China; Information Processing Laboratory, School of Information Engineering, Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, 014010, China.
| |
Collapse
|
241
|
Zhao Y, Qin F, Han S, Li S, Zhao Y, Wang H, Tian J, Cen X. MicroRNAs in drug addiction: Current status and future perspectives. Pharmacol Ther 2022; 236:108215. [DOI: 10.1016/j.pharmthera.2022.108215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/21/2022]
|
242
|
Park K, Clare K, Volkow ND, Pan Y, Du C. Cocaine's effects on the reactivity of the medial prefrontal cortex to ventral tegmental area stimulation: optical imaging study in mice. Addiction 2022; 117:2242-2253. [PMID: 35293056 PMCID: PMC9801493 DOI: 10.1111/add.15869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 02/18/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS The prefrontal cortex (PFC) is modulated by dopaminergic and glutamatergic neurons that project from the ventral tegmental area (VTA) and disruption of this modulation might facilitate impulsive behaviors during cocaine intoxication. Here, we assessed the effects of acute cocaine (30 mg/kg, i.p.) on the reactivity of the PFC to VTA stimulation. METHODS Using a genetically encoded calcium indicator (GCaMP6f), we optically imaged the neuronal Ca2+ reactance in medial PFC (mPFC) in response to 'tonic-like' (5 Hz) and 'phasic-like' (50 Hz) electrical VTA stimulation. The high temporal and spatial resolutions of our optical system allowed us to capture single Ca2+ neuronal transients from individual stimuli with 'tonic-like' stimulation and to visualize single neuronal activation evoked by 'phasic-like' VTA stimulation. RESULTS 'Tonic-like' VTA stimulation induced a rapid increase in mean neuronal Ca2+ in mPFC followed by a plateau and recovery upon termination of stimulation. After cocaine, the mPFC sensitivity to 'tonic-like' VTA stimulation was attenuated, with a 50.4% reduction (P = 0.03) in the number of Ca2+ transients corresponding to single electrical stimuli but the recovery time was lengthened (4.30 ± 0.25 sec to 5.41 ± 0.24 sec, P = 0.03). 'Phasic-like' stimulation evoked a rapid Ca2+ fluorescence increase in mPFC with an immediate decay process, and while cocaine did not affect the peak response (7.17 ± 1.07% versus 7.13 ± 0.96%, P = 0.98) it shortened the recovery time to baseline (3.27 ± 0.11 sec versus 2.38 ± 0.23 sec, P = 0.005). CONCLUSIONS Acute cocaine impairs reactivity of medial prefrontal cortex (mPFC) to ventral tegmental area stimulation, decreasing its sensitivity to 'tonic-like' stimulation and lengthening the recovery time to return to baseline while shortening it for phasic stimulation. These changes in mPFC might contribute to cocaine binging during intoxication.
Collapse
Affiliation(s)
- Kicheon Park
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Kevin Clare
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | | | - Yingtian Pan
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Congwu Du
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
243
|
“Returning to Ordinary Citizenship”: A Qualitative Study of Chinese PWUD’s Self-Management Strategies and Disengagement Model of Identity. Behav Sci (Basel) 2022; 12:bs12080258. [PMID: 36004829 PMCID: PMC9404964 DOI: 10.3390/bs12080258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
How PWUD (people who use drugs) live under drug governance is an important research question. This study adopts a qualitative research method to explore how PWUD in China self-manage after perceiving the dilemma of incomplete citizenship and the social pressure brought by drug control arrangements. Through analysis of 130 PWUD’s files and in-depth interviews with 10 interviewees (from the 24 preliminary interviews), this study found that PWUD developed action strategies of hidden mobility (spatial isolation), disconnection of past experiences (time isolation), instrumental actions, as well as narrative strategies of reframing themselves as ordinary citizens with attempts of reversing identity disadvantages. Further, PWUD’s self-management strategies manifest as a disengagement model in which the actors (PWUD, not rehabilitation agencies) do not intend to develop integrative positive identities through dispersed, practiced behavioral strategies, but attempt to return to pre-addiction, non-socially exclusionary citizenship experiences. The disengagement model and its negative effect on PWUD’s social integration help us reflect on the current implementation of rehabilitation projects and institutional settings of drug governance.
Collapse
|
244
|
Maternal immune activation and adolescent alcohol exposure increase alcohol drinking and disrupt cortical-striatal-hippocampal oscillations in adult offspring. Transl Psychiatry 2022; 12:288. [PMID: 35859084 PMCID: PMC9300672 DOI: 10.1038/s41398-022-02065-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/21/2022] [Accepted: 07/07/2022] [Indexed: 11/08/2022] Open
Abstract
Maternal immune activation (MIA) is strongly associated with an increased risk of developing mental illness in adulthood, which often co-occurs with alcohol misuse. The current study aimed to begin to determine whether MIA, combined with adolescent alcohol exposure (AE), could be used as a model with which we could study the neurobiological mechanisms behind such co-occurring disorders. Pregnant Sprague-Dawley rats were treated with polyI:C or saline on gestational day 15. Half of the offspring were given continuous access to alcohol during adolescence, leading to four experimental groups: controls, MIA, AE, and Dual (MIA + AE). We then evaluated whether MIA and/or AE alter: (1) alcohol consumption; (2) locomotor behavior; and (3) cortical-striatal-hippocampal local field potentials (LFPs) in adult offspring. Dual rats, particularly females, drank significantly more alcohol in adulthood compared to all other groups. MIA led to reduced locomotor behavior in males only. Using machine learning to build predictive models from LFPs, we were able to differentiate Dual rats from control rats and AE rats in both sexes, and Dual rats from MIA rats in females. These data suggest that Dual "hits" (MIA + AE) increases substance use behavior and disrupts activity in reward-related circuits, and that this may be a valuable heuristic model we can use to study the neurobiological underpinnings of co-occurring disorders. Our future work aims to extend these findings to other addictive substances to enhance the translational relevance of this model, as well as determine whether amelioration of these circuit disruptions can reduce substance use behavior.
Collapse
|
245
|
Witek K, Wydra K, Filip M. A High-Sugar Diet Consumption, Metabolism and Health Impacts with a Focus on the Development of Substance Use Disorder: A Narrative Review. Nutrients 2022; 14:2940. [PMID: 35889898 PMCID: PMC9323357 DOI: 10.3390/nu14142940] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 01/01/2023] Open
Abstract
Carbohydrates are important macronutrients in human and rodent diet patterns that play a key role in crucial metabolic pathways and provide the necessary energy for proper body functioning. Sugar homeostasis and intake require complex hormonal and nervous control to proper body energy balance. Added sugar in processed food results in metabolic, cardiovascular, and nervous disorders. Epidemiological reports have shown enhanced consumption of sweet products in children and adults, especially in reproductive age and in pregnant women, which can lead to the susceptibility of offspring's health to diseases in early life or in adulthood and proneness to mental disorders. In this review, we discuss the impacts of high-sugar diet (HSD) or sugar intake during the perinatal and/or postnatal periods on neural and behavioural disturbances as well as on the development of substance use disorder (SUD). Since several emotional behavioural disturbances are recognized as predictors of SUD, we also present how HSD enhances impulsive behaviour, stress, anxiety and depression. Apart from the influence of HSD on these mood disturbances, added sugar can render food addiction. Both food and addictive substances change the sensitivity of the brain rewarding neurotransmission signalling. The results of the collected studies could be important in assessing sugar intake, especially via maternal dietary patterns, from the clinical perspective of SUD prevention or pre-existing emotional disorders. Methodology: This narrative review focuses on the roles of a high-sugar diet (HSD) and added sugar in foods and on the impacts of glucose and fructose on the development of substance use disorder (SUD) and on the behavioural predictors of drugs abuse. The literature was reviewed by two authors independently according to the topic of the review. We searched the PubMed and Scopus databases and Multidisciplinary Digital Publishing Institute open access scientific journals using the following keyword search strategy depending on the theme of the chapter: "high-sugar diet" OR "high-carbohydrate diet" OR "sugar" OR "glucose" OR "fructose" OR "added sugar" AND keywords. We excluded inaccessible or pay-walled articles, abstracts, conference papers, editorials, letters, commentary, and short notes. Reviews, experimental studies, and epidemiological data, published since 1990s, were searched and collected depending on the chapter structure. After the search, all duplicates are thrown out and full texts were read, and findings were rescreened. After the selection process, appropriate papers were included to present in this review.
Collapse
Affiliation(s)
| | | | - Małgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (K.W.); (K.W.)
| |
Collapse
|
246
|
Kobayashi NHC, Farias SV, Luz DA, Machado-Ferraro KM, da Conceição BC, da Silveira CCM, Fernandes LMP, Cartágenes SDC, Ferreira VMM, Fontes-Júnior EA, Maia CDSF. Ketamine plus Alcohol: What We Know and What We Can Expect about This. Int J Mol Sci 2022; 23:ijms23147800. [PMID: 35887148 PMCID: PMC9323326 DOI: 10.3390/ijms23147800] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 01/02/2023] Open
Abstract
Drug abuse has become a public health concern. The misuse of ketamine, a psychedelic substance, has increased worldwide. In addition, the co-abuse with alcohol is frequently identified among misusers. Considering that ketamine and alcohol share several pharmacological targets, we hypothesize that the consumption of both psychoactive substances may synergically intensify the toxicological consequences, both under the effect of drugs available in body systems and during withdrawal. The aim of this review is to examine the toxicological mechanisms related to ketamine plus ethanol co-abuse, as well the consequences on cardiorespiratory, digestive, urinary, and central nervous systems. Furthermore, we provide a comprehensive discussion about the probable sites of shared molecular mechanisms that may elicit additional hazardous effects. Finally, we highlight the gaps of knowledge in this area, which deserves further research.
Collapse
Affiliation(s)
- Natalia Harumi Correa Kobayashi
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Sarah Viana Farias
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Diandra Araújo Luz
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Kissila Márvia Machado-Ferraro
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Brenda Costa da Conceição
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Cinthia Cristina Menezes da Silveira
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Luanna Melo Pereira Fernandes
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Sabrina de Carvalho Cartágenes
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Vânia Maria Moraes Ferreira
- Laboratory of Psychobiology, Psychology Institute, University of Brasília, Campus Universitário Darcy Ribeiro—Asa Norte, Brasília 70910900, DF, Brazil;
| | - Enéas Andrade Fontes-Júnior
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
| | - Cristiane do Socorro Ferraz Maia
- Laboratory of Pharmacology of Inflammation and Behavior, Faculty of Pharmacy, Institute of Health Science, Federal University of Pará, Belém 66075110, PA, Brazil; (N.H.C.K.); (S.V.F.); (D.A.L.); (K.M.M.-F.); (B.C.d.C.); (C.C.M.d.S.); (L.M.P.F.); (S.d.C.C.); (E.A.F.-J.)
- Correspondence: ; Tel.: +55-91-3201-7201
| |
Collapse
|
247
|
Shyu C, Chavez S, Boileau I, Foll BL. Quantifying GABA in Addiction: A Review of Proton Magnetic Resonance Spectroscopy Studies. Brain Sci 2022; 12:918. [PMID: 35884725 PMCID: PMC9316447 DOI: 10.3390/brainsci12070918] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/04/2023] Open
Abstract
Gamma-aminobutyric acid (GABA) signaling plays a crucial role in drug reward and the development of addiction. Historically, GABA neurochemistry in humans has been difficult to study due to methodological limitations. In recent years, proton magnetic resonance spectroscopy (1H-MRS, MRS) has emerged as a non-invasive imaging technique that can detect and quantify human brain metabolites in vivo. Novel sequencing and spectral editing methods have since been developed to allow for quantification of GABA. This review outlines the clinical research utilization of 1H-MRS in understanding GABA neurochemistry in addiction and summarizes current literature that reports GABA measurements by MRS in addiction. Research on alcohol, nicotine, cocaine, and cannabis addiction all suggest medications that modulate GABA signaling may be effective in reducing withdrawal, craving, and other addictive behaviors. Thus, we discuss how improvements in current MRS techniques and design can optimize GABA quantification in future studies and explore how monitoring changes to brain GABA could help identify risk factors, improve treatment efficacy, further characterize the nature of addiction, and provide crucial insights for future pharmacological development.
Collapse
Affiliation(s)
- Claire Shyu
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON M5S 2S1, Canada;
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sofia Chavez
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Isabelle Boileau
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
| | - Bernard Le Foll
- Translational Addiction Research Laboratory, Centre for Addiction and Mental Health, Toronto, ON M5S 2S1, Canada;
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON M5T 1R8, Canada; (S.C.); (I.B.)
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
- Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute, Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, Division of Brain and Therapeutics, University of Toronto, Toronto, ON M5T 1R8, Canada
- Centre for Addiction and Mental Health, Concurrent Outpatient Medical & Psychosocial Addiction Support Services, Toronto, ON M6J 1H4, Canada
- Centre for Addiction and Mental Health, Acute Care Program, Toronto, ON M6J 1H3, Canada
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
- Waypoint Centre for Mental Health Care, Waypoint Research Institute, 500 Church Street, Penetanguishene, ON L9M 1G3, Canada
| |
Collapse
|
248
|
Abstract
When does repeated behaviour constitute behavioural addiction? There has been considerable debate about non-substance-related addictions and how to determine when impaired control over a behaviour is addiction. There are public health benefits to identifying new behavioural addictions if intervention can improve outcomes. However, criteria for establishing new behavioural addictions must guard against diagnostic inflation and the pathologizing of normal problems of living. Criteria should include clinical relevance (Criterion 1), alignment with addiction phenomenology (Criterion 2) and theory (Criterion 3), and taxonomic plausibility (Criterion 4). Against such criteria, evidence does not yet support classification of pornography-use and buying-shopping disorders as addictions.
Collapse
Affiliation(s)
- Matthew J. Gullo
- National Centre for Youth Substance Use Research, The University of Queensland, Brisbane, QLD 4072, Australia
- Corresponding author. E-mail:
| | - Andrew P. Wood
- School of Health & Behavioural Sciences, The University of the Sunshine Coast, Birtinya, QLD 4575, Australia
| | - John B. Saunders
- National Centre for Youth Substance Use Research, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
249
|
Erdoğan A, Özge B, Kopan M, Aktan Y, Kulaksızoğlu B. Second to fourth digit ratio (2D:4D), impulsivity and aggression levels in patients with opiate use disorder and alcohol use disorder. JOURNAL OF SUBSTANCE USE 2022. [DOI: 10.1080/14659891.2022.2098850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Ali Erdoğan
- Department of Psychiatry, Akdeniz University Medical School, Antalya, Turkey
| | - Başak Özge
- Department of Psychiatry, Akdeniz University Medical School, Antalya, Turkey
| | - Metehan Kopan
- Department of Psychiatry, Akdeniz University Medical School, Antalya, Turkey
| | - Yiğitcan Aktan
- Department of Psychiatry, Akdeniz University Medical School, Antalya, Turkey
| | - Burak Kulaksızoğlu
- Department of Psychiatry, Akdeniz University Medical School, Antalya, Turkey
| |
Collapse
|
250
|
Seemiller LR, Logue SF, Gould TJ. Inbred mouse strain differences in alcohol and nicotine addiction-related phenotypes from adolescence to adulthood. Pharmacol Biochem Behav 2022; 218:173429. [PMID: 35820468 PMCID: PMC11524176 DOI: 10.1016/j.pbb.2022.173429] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/18/2022] [Accepted: 07/06/2022] [Indexed: 11/20/2022]
Abstract
Understanding the genetic basis of a predisposition for nicotine and alcohol use across the lifespan is important for public health efforts because genetic contributions may change with age. However, parsing apart subtle genetic contributions to complex human behaviors is a challenge. Animal models provide the opportunity to study the effects of genetic background and age on drug-related phenotypes, while controlling important experimental variables such as amount and timing of drug exposure. Addiction research in inbred, or isogenic, mouse lines has demonstrated genetic contributions to nicotine and alcohol abuse- and addiction-related behaviors. This review summarizes inbred mouse strain differences in alcohol and nicotine addiction-related phenotypes including voluntary consumption/self-administration, initial sensitivity to the drug as measured by sedative, hypothermic, and ataxic effects, locomotor effects, conditioned place preference or place aversion, drug metabolism, and severity of withdrawal symptoms. This review also discusses how these alcohol and nicotine addiction-related phenotypes change from adolescence to adulthood.
Collapse
Affiliation(s)
- Laurel R Seemiller
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Sheree F Logue
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA
| | - Thomas J Gould
- Department of Biobehavioral Health, Penn State University, University Park, PA, USA.
| |
Collapse
|