1
|
Iyer V, Saberi SA, Pacheco R, Sizemore EF, Stockman S, Kulkarni A, Cantwell L, Thakur GA, Hohmann AG. Negative allosteric modulation of CB 1 cannabinoid receptor signaling suppresses opioid-mediated tolerance and withdrawal without blocking opioid antinociception. Neuropharmacology 2024; 257:110052. [PMID: 38936657 PMCID: PMC11261750 DOI: 10.1016/j.neuropharm.2024.110052] [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: 01/07/2024] [Revised: 06/02/2024] [Accepted: 06/21/2024] [Indexed: 06/29/2024]
Abstract
The direct blockade of CB1 cannabinoid receptors produces therapeutic effects as well as adverse side-effects that limit their clinical potential. CB1 negative allosteric modulators (NAMs) represent an indirect approach to decrease the affinity and/or efficacy of orthosteric cannabinoid ligands or endocannabinoids at CB1. We recently reported that GAT358, a CB1-NAM, blocked opioid-induced mesocorticolimbic dopamine release and reward via a CB1-allosteric mechanism of action. Whether a CB1-NAM dampens opioid-mediated therapeutic effects such as analgesia or alters other unwanted opioid side-effects remain unknown. Here, we characterized the effects of GAT358 on nociceptive behaviors in the presence and absence of morphine in male rats. We examined the impact of GAT358 on formalin-evoked pain behavior and Fos protein expression, a marker of neuronal activation, in the lumbar spinal cord. We also assessed the impact of GAT358 on morphine-induced slowing of colonic transit, tolerance, and withdrawal behaviors in male mice. GAT358 attenuated morphine antinociceptive tolerance without blocking acute antinociception and reduced morphine-induced slowing of colonic motility without impacting fecal boli production. GAT358 also produced antinociception in the presence and absence of morphine in the formalin model of inflammatory nociception and reduced the number of formalin-evoked Fos protein-like immunoreactive cells in the lumbar spinal cord. Finally, GAT358 mitigated the somatic signs of naloxone-precipitated, but not spontaneous, opioid withdrawal following chronic morphine dosing. Our results support the therapeutic potential of CB1-NAMs as novel drug candidates aimed at preserving opioid-mediated analgesia while preventing their unwanted side-effects. Our studies also uncover previously unrecognized antinociceptive properties associated with an arrestin-biased CB1-NAM.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Shahin A Saberi
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Romario Pacheco
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Emily Fender Sizemore
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Sarah Stockman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Andrea G Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA; Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
2
|
Rakela S, Sortman BW, Gobin C, Hao S, Caceres-Brun D, Warren BL. Self-administration acquisition latency predicts locomotor sensitivity to cocaine in male rats. Behav Brain Res 2024; 473:115170. [PMID: 39084564 DOI: 10.1016/j.bbr.2024.115170] [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: 05/31/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Individual differences in drug use emerge soon after initial exposure, and only a fraction of individuals who initiate drug use go on to develop a substance use disorder. Variability in vulnerability to establishing drug self-administration behavior is also evident in preclinical rodent models. Latent characteristics that underlie this variability and the relationship between early drug use patterns and later use remain unclear. Here, we attempt to determine whether propensity to establish cocaine self-administration is related to subsequent cocaine self-administration behavior in male Sprague-Dawley rats (n = 14). Prior to initiating training, we evaluated basal locomotor and anxiety-like behavior in a novel open field test. We then trained rats to self-administer cocaine in daily 3 h cocaine (0.75 mg/kg/infusion) self-administration sessions until acquisition criteria (≥30 active lever presses with ≥70 % responding on the active lever in one session) was met and divided rats into Early and Late groups by median-split analysis based on their latency to meet acquisition criteria. After each rat met acquisition criteria, we gave them 10 additional daily cocaine self-administration sessions. We then conducted a progressive ratio, cocaine-induced locomotor sensitivity test, and non-reinforced cocaine seeking test after two weeks of forced abstinence. Early Learners exhibited significantly less locomotion after an acute injection of cocaine, but the groups did not differ in any other behavioral parameter examined. These results indicate that cocaine self-administration acquisition latency is not predictive of subsequent drug-taking behavior, but may be linked to physiological factors like drug sensitivity that can predispose rats to learn the operant task.
Collapse
Affiliation(s)
- Samantha Rakela
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Bo W Sortman
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Christina Gobin
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Sophie Hao
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States
| | - Delfina Caceres-Brun
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States.
| | - Brandon L Warren
- Department of Pharmacodynamics, University of Florida, 1345 Center Dr., Gainesville, FL 32610, United States
| |
Collapse
|
3
|
Schilling L, Singleton SP, Tozlu C, Hédo M, Zhao Q, Pohl KM, Jamison K, Kuceyeski A. Sex-specific differences in brain activity dynamics of youth with a family history of substance use disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.610959. [PMID: 39282344 PMCID: PMC11398379 DOI: 10.1101/2024.09.03.610959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
An individual's risk of substance use disorder (SUD) is shaped by a complex interplay of potent biosocial factors. Current neurodevelopmental models posit vulnerability to SUD in youth is due to an overreactive reward system and reduced inhibitory control. Having a family history of SUD is a particularly strong risk factor, yet few studies have explored its impact on brain function and structure prior to substance exposure. Herein, we utilized a network control theory approach to quantify sex-specific differences in brain activity dynamics in youth with and without a family history of SUD, drawn from a large cohort of substance-naïve youth from the Adolescent Brain Cognitive Development Study. We summarize brain dynamics by calculating transition energy, which probes the ease with which a whole brain, region or network drives the brain towards a specific spatial pattern of activation (i.e., brain state). Our findings reveal that a family history of SUD is associated with alterations in the brain's dynamics wherein: i) independent of sex, certain regions' transition energies are higher in those with a family history of SUD and ii) there exist sex-specific differences in SUD family history groups at multiple levels of transition energy (global, network, and regional). Family history-by-sex effects reveal that energetic demand is increased in females with a family history of SUD and decreased in males with a family history of SUD, compared to their same-sex counterparts with no SUD family history. Specifically, we localize these effects to higher energetic demands of the default mode network in females with a family history of SUD and lower energetic demands of attention networks in males with a family history of SUD. These results suggest a family history of SUD may increase reward saliency in males and decrease efficiency of top-down inhibitory control in females. This work could be used to inform personalized intervention strategies that may target differing cognitive mechanisms that predispose individuals to the development of SUD.
Collapse
Affiliation(s)
- Louisa Schilling
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | | | - Ceren Tozlu
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Marie Hédo
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Qingyu Zhao
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Kilian M Pohl
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Keith Jamison
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Amy Kuceyeski
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
4
|
Hagarty DP, Dawoud A, Brea Guerrero A, Phillips K, Strong CE, Jennings SD, Crawford M, Martinez K, Csernecky O, Saland SK, Kabbaj M. Exploring ketamine's reinforcement, cue-induced reinstatement, and nucleus accumbens cFos activation in male and female long evans rats. Neuropharmacology 2024; 255:110008. [PMID: 38797243 DOI: 10.1016/j.neuropharm.2024.110008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/26/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024]
Abstract
Ketamine (KET), a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, has rapid onset of antidepressant effects in Treatment-Resistant Depression patients and repeated infusions are required to sustain its antidepressant properties. However, KET is an addictive drug, and so more preclinical and clinical research is needed to assess the safety of recurring treatments in both sexes. Thus, the aim of this study was to investigate the reinforcing properties of various doses of KET (0-, 0.125-, 0.25-, 0.5 mg/kg/infusion) and assess KET's cue-induced reinstatement and neuronal activation in both sexes of Long Evans rats. Neuronal activation was assessed using the protein expression of the immediate early gene cFos in the nucleus accumbens (Nac), an important brain area implicated in reward, reinforcement and reinstatement to most drug-related cues. Our findings show that KET has reinforcing effects in both male and female rats, albeit exclusively at the highest two doses (0.25 and 0.5 mg/kg/infusion). Furthermore, we noted sex differences, particularly at the highest dose of ketamine, with female rats displaying a higher rate of self-administration. Interestingly, all groups that self-administered KET reinstated to drug-cues. Following drug cue-induced reinstatement test in rats exposed to KET (0.25 mg/kg/infusion) or saline, there was higher cFos protein expression in KET-treated animals compared to saline controls, and higher cFos expression in the core compared to the shell subregions of the Nac. As for reinstatement, there were no notable sex differences reported for cFos expression in the Nac. These findings reveal some sex and dose dependent effects in KET's reinforcing properties and that KET at all doses induced similar reinstatement in both sexes. This study also demonstrated that cues associated with ketamine induce comparable neuronal activation in the Nac of both male and female rats. This work warrants further research into the potential addictive properties of KET, especially when administered at lower doses which are now being used in the clinic for treating various psychopathologies.
Collapse
Affiliation(s)
- Devin P Hagarty
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Adam Dawoud
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Alfonso Brea Guerrero
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Kaynas Phillips
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Caroline E Strong
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Sarah Dollie Jennings
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Michelle Crawford
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Katherine Martinez
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Olivia Csernecky
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Samantha K Saland
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, College of Medicine, Program in Neuroscience, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
5
|
Heidari A, Hajikarim-Hamedani A, Hosseindoost S, Ghane Y, Sadat-Shirazi M, Zarrindast MR. Parental Exposure to Morphine Before Conception Decreases Morphine and Cocaine-Induced Locomotor Sensitization in Male Offspring. Dev Psychobiol 2024; 66:e22514. [PMID: 38922890 DOI: 10.1002/dev.22514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
Repeated exposure to abused drugs leads to reorganizing synaptic connections in the brain, playing a pivotal role in the relapse process. Additionally, recent research has highlighted the impact of parental drug exposure before gestation on subsequent generations. This study aimed to explore the influence of parental morphine exposure 10 days prior to pregnancy on drug-induced locomotor sensitization. Adult male and female Wistar rats were categorized into morphine-exposed and control groups. Ten days after their last treatment, they were mated, and their male offspring underwent morphine, methamphetamine, cocaine, and nicotine-induced locomotor sensitization tests. The results indicated increased locomotor activity in both groups after drug exposure, although the changes were attenuated in morphine and cocaine sensitization among the offspring of morphine-exposed parents (MEPs). Western blotting analysis revealed altered levels of D2 dopamine receptors (D2DRs) in the prefrontal cortex and nucleus accumbens of the offspring from MEPs. Remarkably, despite not having direct in utero drug exposure, these offspring exhibited molecular alterations affecting morphine and cocaine-induced sensitization. The diminished sensitization to morphine and cocaine suggested the development of a tolerance phenotype in these offspring. The changes in D2DR levels in the brain might play a role in these adaptations.
Collapse
Affiliation(s)
- Amirhossein Heidari
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Saereh Hosseindoost
- Pain Research Center, Neuroscience Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Yekta Ghane
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mitra Sadat-Shirazi
- Development, Molecular & Chemical Biology, Tufts University, Boston, Massachusetts, USA
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Sardari M, Mohammadpourmir F, Hosseinzadeh Sahafi O, Rezayof A. Neuronal biomarkers as potential therapeutic targets for drug addiction related to sex differences in the brain: Opportunities for personalized treatment approaches. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111068. [PMID: 38944334 DOI: 10.1016/j.pnpbp.2024.111068] [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: 11/25/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/01/2024]
Abstract
Biological sex disparities manifest at various stages of drug addiction, including craving, substance abuse, abstinence, and relapse. These discrepancies are underpinned by notable distinctions in neurobiological substrates, encompassing brain structures, functions, and neurotransmitter systems implicated in drug addiction. Neuronal biomarkers, such as neurotransmitters, signaling proteins, and genes may be associated with the diagnosis, prognosis, and treatment outcomes in both biological sexes afflicted by drug abuse. Sex differences in the neural reward system, mainly through dopaminergic transmission during drug abuse, can be attributed to modifications in neurotransmitter systems and signaling pathways. This results in distinct patterns of neural activation and responsiveness to addictive substances in males and females. Sex hormones, the estrus/menstrual cycle, and cerebral neurochemistry contribute to the progression of psychological and physiological dependence in both male and female individuals grappling with addiction. Moreover, the alteration of sex hormone balance and neurotransmitter release plays a pivotal role in substance use disorders, subsequently modulating cognitive functions pertinent to reward, including memory formation, decision-making, and locomotor activity. Comparative investigations reveal distinctions in brain region volume, gene expression, neuronal firing, and circuitry in substance use disorders affecting individuals of both biological sexes. This review examines prevalent substance use disorders to elucidate the impact of sex hormones as therapeutic biomarkers on the mesocorticolimbic neurotransmitter systems via diverse mechanisms within the addicted brain. We underscore the imperative necessity of considering these variations to gain a deeper comprehension of addiction mechanisms and potentially discern sex-specific neuronal biomarkers for tailored therapeutic interventions.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Farina Mohammadpourmir
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Oveis Hosseinzadeh Sahafi
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| |
Collapse
|
7
|
Krolick KN, Cao J, Gulla EM, Bhardwaj M, Marshall SJ, Zhou EY, Kiss AJ, Choueiry F, Zhu J, Shi H. Subregion-specific transcriptomic profiling of rat brain reveals sex-distinct gene expression impacted by adolescent stress. Neuroscience 2024; 553:19-39. [PMID: 38977070 PMCID: PMC11444371 DOI: 10.1016/j.neuroscience.2024.07.002] [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: 01/18/2024] [Revised: 05/14/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Stress during adolescence clearly impacts brain development and function. Sex differences in adolescent stress-induced or exacerbated emotional and metabolic vulnerabilities could be due to sex-distinct gene expression in hypothalamic, limbic, and prefrontal brain regions. However, adolescent stress-induced whole-genome expression changes in key subregions of these brain regions were unclear. In this study, female and male adolescent Sprague Dawley rats received one-hour restraint stress daily from postnatal day (PD) 32 to PD44. Corticosterone levels, body weights, food intake, body composition, and circulating adiposity and sex hormones were measured. On PD44, brain and blood samples were collected. Using RNA-sequencing, sex-specific differences in stress-induced differentially expressed (DE) genes were identified in subregions of the hypothalamus, limbic system, and prefrontal cortex. Canonical pathways reflected well-known sex-distinct maladies and diseases, substantiating the therapeutic potential of the DE genes found in the current study. Thus, we proposed specific sex distinct, adolescent stress-induced transcriptional changes found in the current study as examples of the molecular bases for sex differences witnessed in stress induced or exacerbated emotional and metabolic disorders. Future behavioral studies and single-cell studies are warranted to test the implications of the DE genes identified in this study in sex-distinct stress-induced susceptibilities.
Collapse
Affiliation(s)
| | - Jingyi Cao
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Evelyn M Gulla
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Meeta Bhardwaj
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | | | - Ethan Y Zhou
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Andor J Kiss
- Center for Bioinformatics & Functional Genomics, Miami University, Oxford, OH 45056, USA.
| | - Fouad Choueiry
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA.
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Haifei Shi
- Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
8
|
Cong Z, Yang L, Zhao Z, Zheng G, Bao C, Zhang P, Wang J, Zheng W, Yao Z, Hu B. Disrupted dynamic brain functional connectivity in male cocaine use disorder: Hyperconnectivity, strongly-connected state tendency, and links to impulsivity and borderline traits. J Psychiatr Res 2024; 176:218-231. [PMID: 38889552 DOI: 10.1016/j.jpsychires.2024.06.012] [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: 11/19/2023] [Revised: 05/28/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024]
Abstract
Cocaine use is a major public health problem with serious negative consequences at both the individual and societal levels. Cocaine use disorder (CUD) is associated with cognitive and emotional impairments, often manifesting as alterations in brain functional connectivity (FC). This study employed resting-state functional magnetic resonance imaging (rs-fMRI) to examine dynamic FC in 38 male participants with CUD and 31 matched healthy controls. Using group spatial independent component analysis (group ICA) combined with sliding window approach, we identified two recurring distinct connectivity states: the strongly-connected state (state 1) and weakly-connected state (state 2). CUD patients exhibited significant increased mean dwell and fraction time in state 1, and increased transitions from state 2 to state 1, demonstrated significant strongly-connected state tendency. Our analysis revealed abnormal FC patterns that are state-dependent and state-shared in CUD patients. This study observed hyperconnectivity within the default mode network (DMN) and between DMN and other networks, which varied depending on the state. Furthermore, after adjustment for multiple comparisons, we found significant correlations between these altered dynamic FCs and clinical measures of impulsivity and borderline personality disorder. The disrupted FC and repetitive effects of precuneus and angular gyrus across correlations suggested that they might be the important hub of neural circuits related behaviorally and mentally in CUD. In summary, our study highlighted the potential of these disrupted FC as neuroimaging biomarkers and therapeutic targets, and provided new insights into the understanding of the neurophysiologic mechanisms of CUD.
Collapse
Affiliation(s)
- Zhaoyang Cong
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China; State Key Laboratory of Digital Medical Engineering, School of Instrument Science and Engineering, Southeast University, Nanjing, 210096, China
| | - Lin Yang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Ziyang Zhao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Guowei Zheng
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China; School of Computer Science and Technology, Harbin Institute of Technology, Harbin, 150006, China
| | - Cong Bao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pengfei Zhang
- Second Clinical School, Lanzhou University, Lanzhou, 730000, China
| | - Jun Wang
- Second Clinical School, Lanzhou University, Lanzhou, 730000, China
| | - Weihao Zheng
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Zhijun Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China.
| | - Bin Hu
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, Lanzhou, 730000, China; School of Medical Technology, Beijing Institute of Technology, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China; Joint Research Center for Cognitive Neurosensor Technology of Lanzhou University & Institute of Semiconductors, Chinese Academy of Sciences, China.
| |
Collapse
|
9
|
Hrelja KM, Kawkab C, Avramidis DK, Ramaiah S, Winstanley CA. Increased risky choice during forced abstinence from fentanyl on the cued rat gambling task. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06659-w. [PMID: 39078498 DOI: 10.1007/s00213-024-06659-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/23/2024] [Indexed: 07/31/2024]
Abstract
RATIONALE The use of illicit opioids has arguably never been more risky; street drug potency can be dangerously high, is often unknown to the consumer, and results in multiple daily fatalities worldwide. Furthermore, substance use disorder (SUD) is associated with increased maladaptive, risky decisions in laboratory-based gambling tasks. Animal studies can help determine whether this decision-making deficit is a cause or consequence of drug use. However, most experiments have only assessed psychostimulant drugs. OBJECTIVES To assess differences in decision-making strategies both before, during, and after self-administration of fentanyl in male and female Long Evans rats. METHODS Male and female Long Evans rats were trained to perform the rat gambling task (rGT), loosely based on the Iowa Gambling Task (IGT) used clinically, and/or self-administer fentanyl. We used the cued version of the rGT, in which sound and light stimuli signal sugar pellet rewards, as cocaine self-administration has the greatest effects on decision making in this task variant. RESULTS After training on the cued rGT, female rats self-administered fentanyl more readily, an effect that was most apparent in optimal decision-makers. Contrary to previous reports using cocaine self-administration, decision-making was unaffected during fentanyl self-administration training in either sex. However, risky decision-making increased throughout forced abstinence from fentanyl in males. CONCLUSIONS These findings complement those from human subjects, in whom preference for uncertain outcomes increased before relapse. These data highlight an abstinence-induced change in cognition that is unique to opiates as compared to psychostimulants, and which may critically contribute to the maintenance of addiction and relapse.
Collapse
Affiliation(s)
- Kelly M Hrelja
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| | - Carol Kawkab
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Dimitrios K Avramidis
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Shrishti Ramaiah
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Catharine A Winstanley
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
10
|
Kocum CG, Cam Y, Shay DA, Schweizer TA, Konrad ER, Houska TK, Sardina CA, Schachtman TR, Vieira-Potter VJ, Will MJ. Voluntary wheel running access produces opposite effects in male and female rats on both palatable diet consumption and associated ventral striatal opioid- and dopamine-related gene expression. Front Integr Neurosci 2024; 18:1426219. [PMID: 39131599 PMCID: PMC11310025 DOI: 10.3389/fnint.2024.1426219] [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: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 08/13/2024] Open
Abstract
The relationship between physical activity levels and feeding behaviors has been a focus of preclinical research for decades, yet this interaction has only recently been explored for potential sex differences. The aim of the present study was to isolate sex-dependent effects of voluntary wheel running (RUN) vs. sedentary locked wheel (SED) home cage conditions on palatability-driven feeding behavior using a 2-diet choice task between standard chow and a high-fat diet. The sex-dependent effects of physical activity on feeding behavior were examined following a within-subject novel reversal design of physical activity conditions (i.e., RUN > SED > RUN), to assess temporal sensitivity of the interaction. Following the final 2 weeks of reestablished and sustained RUN vs. SED conditions in separate groups of both males and females, reward-related opioid and dopamine gene expression within the nucleus accumbens (Acb) brain region were analyzed. Results demonstrated that the initial RUN > SED transition led to sex-dependent effects of SED condition, as males increased, and females decreased their high fat consumption, compared to their respective high fat consumption during previous RUN condition phase. Following reintroduction to the RUN condition, males decreased, and females increased their high fat consumption, compared to their separate SED control group. Last, sex-dependent shifts in ventral striatal opioid- and dopamine-related gene expression were observed to parallel the behavioral effects. The major findings of the study reveal that SED and RUN home cage conditions shift palatability-driven feeding in the opposite direction for males and females, these effects are sensitive to reversal, and these sex-dependent feeding behaviors track sex-dependent changes to critical reward-related gene expression patterns in the Acb. Considering the present high rates of sedentary behavior and obesity, furthering our understanding of the interaction between physical activity (or lack thereof) and feeding behavior should be a priority, especially in the context of these divergent sex-dependent outcomes.
Collapse
Affiliation(s)
- Courtney G. Kocum
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| | - Yonca Cam
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| | - Dusti A. Shay
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Tim A. Schweizer
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
- Department of Biological Sciences, University of Missouri, Columbia, MO, United States
| | - Ella R. Konrad
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| | - Tabitha K. Houska
- Department of Biological Sciences, University of Missouri, Columbia, MO, United States
| | - Carlos A. Sardina
- Department of Philosophy, University of Missouri, Columbia, MO, United States
| | - Todd R. Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| | - Victoria J. Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Matthew J. Will
- Department of Psychological Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
11
|
García-Cabrerizo R, Cryan JF. A gut (microbiome) feeling about addiction: Interactions with stress and social systems. Neurobiol Stress 2024; 30:100629. [PMID: 38584880 PMCID: PMC10995916 DOI: 10.1016/j.ynstr.2024.100629] [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: 11/23/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
In recent years, an increasing attention has given to the intricate and diverse connection of microorganisms residing in our gut and their impact on brain health and central nervous system disease. There has been a shift in mindset to understand that drug addiction is not merely a condition that affects the brain, it is now being recognized as a disorder that also involves external factors such as the intestinal microbiota, which could influence vulnerability and the development of addictive behaviors. Furthermore, stress and social interactions, which are closely linked to the intestinal microbiota, are powerful modulators of addiction. This review delves into the mechanisms through which the microbiota-stress-immune axis may shape drug addiction and social behaviors. This work integrates preclinical and clinical evidence that demonstrate the bidirectional communication between stress, social behaviors, substance use disorders and the gut microbiota, suggesting that gut microbes might modulate social stress having a significance in drug addiction.
Collapse
Affiliation(s)
- Rubén García-Cabrerizo
- IUNICS, University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Medicine, University of the Balearic Islands, Palma, Spain
| | - John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Orndorff M, Shipp GM, Kerver JM, Ondersma SJ, Alshaarawy O. Trends in cocaine use among United States females of reproductive age, 2005-2019. Am J Addict 2024; 33:313-319. [PMID: 37924245 DOI: 10.1111/ajad.13502] [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: 03/27/2023] [Revised: 09/18/2023] [Accepted: 10/21/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The prevalence of cocaine use has increased in the United States, with an estimated 5.5 million people have used it at least once in 2019. We report trends in cocaine use for reproductive age females who participated in the National Survey on Drug Use and Health (NSDUH, 2005-2019). METHODS Interviewers for NSDUH recruited and assessed a representative sample of the US noninstitutionalized population. The prevalence of cocaine use was estimated annually for females (15-44 years; n = 295,751). Joinpoint regression was then used to test for significant changes in trends. RESULTS Approximately 2.4% of females (n = 8136) reported past 12-month cocaine use. Joinpoint regression revealed an initial decline in cocaine use prevalence between 2005 and 2011, followed by a robust increase (2011-2019 annual percent change = 5.2; 95% confidence interval = 2.6, 7.8%). This nonlinear trend was observed for all subgroups, except for adolescent and pregnant females for whom a decrease in cocaine use prevalence was observed. DISCUSSION AND CONCLUSIONS Although the prevalence of cocaine use remains scarce among US females, the uptake in use after a period of initial decline is concerning, highlighting the need for continued public health awareness and action. SCIENTIFIC SIGNIFICANCE Cocaine use among females of reproductive age is particularly concerning given the increased likelihood of transitioning to disordered use and the risk of use during pregnancy. Previous studies have not reported cocaine use estimates specific to reproductive age females or only reported co-drug use patterns, without detail of specific cocaine use trends.
Collapse
Affiliation(s)
- Madelyn Orndorff
- College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Gayle M Shipp
- Charles Stewart Mott Department of Public Health, College of Human Medicine, Michigan State University, Flint, Michigan, USA
| | - Jean M Kerver
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Steven J Ondersma
- Charles Stewart Mott Department of Public Health, College of Human Medicine, Michigan State University, Flint, Michigan, USA
| | - Omayma Alshaarawy
- Department of Family Medicine, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
13
|
Andreu M, Balcells-Olivero M, Alcaraz N, Marco O, Bueno L, Gual A, Barrio P. Destination Matters More: Relapse following Hospital-Based Treatment of Substance Use Disorders With and Without Co-Occurring Disorders. J Dual Diagn 2024; 20:111-121. [PMID: 38367999 DOI: 10.1080/15504263.2024.2311634] [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] [Indexed: 02/19/2024]
Abstract
OBJECTIVES Addressing substance use in psychiatric care encounters significant barriers, but the emergence of specialized services offers an opportunity to advance and scale up the integration of addiction services within psychiatric settings. However, research gaps still exist in this field, particularly in understanding the substance relapse rates of people with co-occurring disorders after a psychiatric hospitalization. This study aimed to investigate and compare the relapse rates of patients under inpatient care with exclusively addiction-related issues and those with co-occurring disorders after a hospitalization in a psychiatric ward and gain insights into differences in outcomes for these two patient groups. METHODS This retrospective analysis examined electronic medical records of patients admitted to the Acute Psychiatry Ward of the Hospital Clinic of Barcelona with a substance use disorder diagnosis between January 2019 and February 2021. Cox regression was used to identify variables independently associated with the first relapse episode. RESULTS From a total of 318 admissions (79.2% with psychiatric comorbidity), 76.1% relapsed during the study follow-up, with a median survival time of 54 days. Younger age, female gender, voluntary admission, and outpatient follow-up were independently associated with relapse. The presence of a co-occurring disorder was not associated with relapse. CONCLUSION This study highlights the need for interventions aimed at improving post-discharge abstinence rates for addiction-related hospitalizations. It also challenges the notion that co-occurring disorders automatically imply a worsened prognosis and emphasizes the importance of addressing addiction and psychiatric comorbidity in a comprehensive, integrated, and specialized manner.
Collapse
Affiliation(s)
- Magalí Andreu
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Doctoral Program of Medicine and Translational Research, University of Barcelona, Barcelona, Spain
| | - Mercè Balcells-Olivero
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Noelia Alcaraz
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Oriol Marco
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Laura Bueno
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Antoni Gual
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Barrio
- Research Group of Clinical Addictions (GRAC-GRE), Department of Psychiatry, Clinical Institute of Neuroscience, Psychiatry and Psychology Service, Hospital Clínic i Universitari de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
14
|
Mahrs-Gould R, Jallo N, Svikis D, Ameringer S, Robins J, Elswick RK. Family history of substance problems among African Americans: Associations with drug use, drug use disorder, and prescription drug misuse. J Ethn Subst Abuse 2024:1-28. [PMID: 38530153 PMCID: PMC11424775 DOI: 10.1080/15332640.2024.2331108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
A family history of substance problems is a well-known risk factor for substance use and use disorders; however, much of this research has been conducted in studies with predominantly White subjects. The aim of this study was to examine the associations between family history density of substance problems and drug use, risk for drug use disorder, and prescription drug misuse in a sample of African American adults. Results indicate that family history density of substance problems increased the risk for all drug outcomes in the full sample. However, when subgroup analyses by gender were conducted, family history was not a risk factor among men for prescription drug misuse.
Collapse
Affiliation(s)
| | - Nancy Jallo
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - Dace Svikis
- Virginia Commonwealth University, Richmond, Virginia, USA
- Institute for Women's Health, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Jo Robins
- Virginia Commonwealth University, Richmond, Virginia, USA
| | - R K Elswick
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
15
|
Lorenzetti V, Gaillard A, McTavish E, Grace S, Rossetti MG, Batalla A, Bellani M, Brambilla P, Chye Y, Conrod P, Cousijn J, Labuschagne I, Clemente A, Mackey S, Rendell P, Solowij N, Suo C, Li CSR, Terrett G, Thompson PM, Yücel M, Garavan H, Roberts CA. Cannabis Dependence is Associated with Reduced Hippocampal Subregion Volumes Independently of Sex: Findings from an ENIGMA Addiction Working Group Multi-Country Study. Cannabis Cannabinoid Res 2024. [PMID: 38498015 DOI: 10.1089/can.2023.0204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Background: Males and females who consume cannabis can experience different mental health and cognitive problems. Neuroscientific theories of addiction postulate that dependence is underscored by neuroadaptations, but do not account for the contribution of distinct sexes. Further, there is little evidence for sex differences in the neurobiology of cannabis dependence as most neuroimaging studies have been conducted in largely male samples in which cannabis dependence, as opposed to use, is often not ascertained. Methods: We examined subregional hippocampus and amygdala volumetry in a sample of 206 people recruited from the ENIGMA Addiction Working Group. They included 59 people with cannabis dependence (17 females), 49 cannabis users without cannabis dependence (20 females), and 98 controls (33 females). Results: We found no group-by-sex effect on subregional volumetry. The left hippocampal cornu ammonis subfield 1 (CA1) volumes were lower in dependent cannabis users compared with non-dependent cannabis users (p<0.001, d=0.32) and with controls (p=0.022, d=0.18). Further, the left cornu ammonis subfield 3 (CA3) and left dentate gyrus volumes were lower in dependent versus non-dependent cannabis users but not versus controls (p=0.002, d=0.37, and p=0.002, d=0.31, respectively). All models controlled for age, intelligence quotient (IQ), alcohol and tobacco use, and intracranial volume. Amygdala volumetry was not affected by group or group-by-sex, but was smaller in females than males. Conclusions: Our findings suggest that the relationship between cannabis dependence and subregional volumetry was not moderated by sex. Specifically, dependent (rather than non-dependent) cannabis use may be associated with alterations in selected hippocampus subfields high in cannabinoid type 1 (CB1) receptors and implicated in addictive behavior. As these data are cross-sectional, it is plausible that differences predate cannabis dependence onset and contribute to the initiation of cannabis dependence. Longitudinal neuroimaging work is required to examine the time-course of the onset of subregional hippocampal alterations in cannabis dependence, and their progression as cannabis dependence exacerbates or recovers over time.
Collapse
Affiliation(s)
- Valentina Lorenzetti
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Alexandra Gaillard
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- Centre for Mental Health and Department of Health Sciences and Biostatistics, Swinburne University, Hawthorn, Australia
| | - Eugene McTavish
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Sally Grace
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Maria Gloria Rossetti
- UOC Psichiatria, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy
| | - Albert Batalla
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Marcella Bellani
- Section of Psychiatry, Department of Neuroscience, Biomedicine and Movement Science, University of Verona, Verona, Italy
| | - Paolo Brambilla
- UOC Psichiatria, Azienda Ospedaliera Universitaria Integrata (AOUI), Verona, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Yann Chye
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Patricia Conrod
- Department of Psychiatry, Université de Montreal, CHU Ste Justine Hospital, Montreal, Canada
| | - Janna Cousijn
- Neuroscience of Addiction Lab, Center for Substance Use and Addiction Research (CESAR), Department of Psychology, Education & Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Izelle Labuschagne
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Adam Clemente
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Scott Mackey
- Department of Psychiatry, University of Vermont, Burlington, Vermont, USA
| | - Peter Rendell
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Nadia Solowij
- School of Psychology, Faculty of the Arts, Social Sciences and Humanities, University of Wollongong, Wollongong, Australia
| | - Chao Suo
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Clayton, Australia
| | - Chiang-Shan R Li
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Gill Terrett
- Neuroscience of Addiction and Mental Health Program, Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - Paul M Thompson
- Department of Neurology, Imaging Genetics Center, Stevens Institute for Neuroimaging & Informatics, Keck School of Medicine, University of Southern California, Marina del Rey, California, USA
| | - Murat Yücel
- QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Hugh Garavan
- School of Psychology, Faculty of Health and Behavioural Sciences, University of Queensland, St Lucia, Australia
| | - Carl A Roberts
- Department of Psychology, Institute of Population Health, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
16
|
Martínez-Caballero MÁ, Calpe-López C, García-Pardo MP, Arenas MC, de la Rubia Ortí JE, Bayona-Babiloni R, Aguilar MA. Behavioural traits related with resilience or vulnerability to the development of cocaine-induced conditioned place preference after exposure of female mice to vicarious social defeat. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110912. [PMID: 38097163 DOI: 10.1016/j.pnpbp.2023.110912] [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: 10/01/2023] [Revised: 11/30/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023]
Abstract
Exposure to stress induced by intermittent repeated social defeat (IRSD) increases vulnerability to the development of cocaine-induced conditioned place preference (CPP) among male mice; however, some defeated mice are resilient to these effects of stress. In the present study we evaluated the effects of vicarious IRSD (VIRSD) in female mice and explored behavioural traits that are potentially predictive of resilience. C57BL/6 female mice (n = 28) were exposed to VIRSD, which consisted of the animals witnessing a short experience of social defeat by a male mouse on postnatal day (PND) 47, 50, 53 and 56. The control group (n = 10) was not exposed to stress. Blood samples were collected on PND 47 and 56 for corticosterone and interleukin-6 determinations. On PND 57-58, female mice performed several behavioural tests (elevated plus maze, hole-board, object recognition, social interaction, TST and splash tests). Three weeks later, the effects of cocaine (1.5 mg/kg) on the CPP paradigm were assessed. VIRSD decreased corticosterone levels (on PND 56), increased interleukin-6 levels, enhanced novelty-seeking, improved recognition memory and induced anxiety- and depression-like symptoms. Control and VIRSD female mice did not acquire CPP, although some stressed individuals with certain behavioural traits - including a high novelty-seeking profile or the development of depression-like behaviour in the splash test shortly after VIRSD - acquired cocaine CPP. Our results confirm that some behavioural traits of female mice are associated with vulnerability or resilience to the long-term effects of social stress on cocaine reward, as previously observed in males.
Collapse
Affiliation(s)
- Maria Ángeles Martínez-Caballero
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behaviour Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Claudia Calpe-López
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behaviour Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain
| | - Maria Pilar García-Pardo
- Department of Psychology and Sociology, Faculty of Social Sciences, University of Zaragoza, Teruel, Spain
| | | | | | - Raquel Bayona-Babiloni
- Department of Basic Medical Sciences, Catholic University of Valencia San Vicente Martir, Valencia, Spain
| | - Maria Asunción Aguilar
- Neurobehavioural Mechanisms and Endophenotypes of Addictive Behaviour Research Unit, Department of Psychobiology, University of Valencia, Valencia, Spain.
| |
Collapse
|
17
|
Recław R, Chmielowiec K, Suchanecka A, Boroń A, Chmielowiec J, Strońska-Pluta A, Kowalski MT, Masiak J, Trybek G, Grzywacz A. The Influence of Genetic Polymorphic Variability of the Catechol-O-methyltransferase Gene in a Group of Patients with a Diagnosis of Behavioural Addiction, including Personality Traits. Genes (Basel) 2024; 15:299. [PMID: 38540358 PMCID: PMC10969953 DOI: 10.3390/genes15030299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/24/2024] [Accepted: 02/24/2024] [Indexed: 04/02/2024] Open
Abstract
Gambling Disorder (GD) is characterised by a harmful, enduring, and recurrent involvement in betting-related behaviours. Therefore, GD shares similar biological mechanisms and symptoms to substance use disorders (SUD). Therefore, in this study, we chose the behavioural addictions group. During the examination and recruitment to the study, it turned out that all the people undergoing treatment for gambling addiction were also addicted to amphetamines, which is consistent with the biological mechanism related to cerebral neurotransmission. The aim of the study was to investigate the association of the COMT gene polymorphism with behavioral addiction. The study group consisted of 307 participants: 107 men with gambling disorder and amphetamine dependency (mean age = 27.51, SD = 5.25) and 200 non-addicted, nor dependent, free from neuro-psychiatric disorders control group men (mean age = 20.20, SD = 4.51). Both groups were subjected to psychometric evaluation using the State-Trait Anxiety Inventory and the NEO Five-Factor Personality Inventory. Genomic DNA was extracted from venous blood following standard protocols. Determination of the rs4680 polymorphism in the COMT gene was performed using the real-time PCR technique. Statistically significant differences in the frequency of rs4680 genotypes were found in the tested sample of subjects compared with the control group (p = 0.03543). Subjects with gambling disorder and amphetamine use disorder compared to the control group obtained higher scores in the assessment of the STAI trait scale (p = 0.0019), state scale (p < 0.0000), and NEO-FFI Neuroticism scale (p < 0.0000). Significantly lower results were obtained for the NEO-FFI Agreeability scale (p < 0.0000). Additionally, a significant statistical impact of gambling disorder and amphetamine use disorder, and the COMT rs4680 genotype was demonstrated for the score of the STAI trait (p = 0.0351) and state (p = 0.0343) and the NEO-FFI Conscientiousness scale (p = 0.0018). We conclude that COMT and its polymorphic variant influence the development of addiction. Still, considering its multifactorial and polygenic nature, it should be combined with other factors such as personality.
Collapse
Affiliation(s)
- Remigiusz Recław
- Foundation Strong in the Spirit, 60 Sienkiewicza St., 90-058 Łódź, Poland;
| | - Krzysztof Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (K.C.); (J.C.)
| | - Aleksandra Suchanecka
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| | - Agnieszka Boroń
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, Aleja Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland;
| | - Jolanta Chmielowiec
- Department of Hygiene and Epidemiology, Collegium Medicum, University of Zielona Góra, 28 Zyty St., 65-046 Zielona Góra, Poland; (K.C.); (J.C.)
| | - Aleksandra Strońska-Pluta
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| | - Michał Tomasz Kowalski
- Clinical Department of Cardiology, Nowa Sól Multidisciplinary Hospital, 67-100 Nowa Sol, Poland;
| | - Jolanta Masiak
- II Department of Psychiatry and Psychiatric Rehabilitation, Medical University of Lublin, 1 Głuska St., 20-059 Lublin, Poland;
| | - Grzegorz Trybek
- Department of Oral Surgery, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland;
- Maxillofacial Surgery Clinic, 4th Military Clinical Hospital in Wroclaw, ul. Rudolfa Weigla 5, 50-981 Wrolaw, Poland
| | - Anna Grzywacz
- Independent Laboratory of Health Promotion, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich 72 St., 70-111 Szczecin, Poland; (A.S.); (A.S.-P.)
| |
Collapse
|
18
|
Menéndez-Delmestre R, Agosto-Rivera JL, González-Segarra AJ, Segarra AC. Cocaine sensitization in male rats requires activation of estrogen receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579327. [PMID: 38370714 PMCID: PMC10871307 DOI: 10.1101/2024.02.07.579327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Gonadal steroids play a modulatory role in cocaine use disorders, and are responsible for many sex differences observed in the behavioral response to cocaine. In females, it is well established that estradiol enhances the behavioral response to cocaine. In males, we have recently shown that testosterone enhances sensitization to cocaine but its mechanism of action remains to be elucidated. The current study investigated the contribution of DHT, a non-aromatizable androgen, and of estradiol, in regulating cocaine-induced sensitization in male rats. Gonadectomized (GDX) male rats treated with estradiol sensitized to repeated cocaine administration, while GDX rats treated with DHT did not, implicating estradiol in cocaine sensitization. Furthermore, intact male rats treated with the antiestrogen ICI 182,780 did not show sensitization to repeated cocaine. This study demonstrates the pivotal role of estradiol in cocaine-induced neuroplasticity and neuroadaptations in the rodent brain.
Collapse
Affiliation(s)
- Raissa Menéndez-Delmestre
- Physiology Department, School of Medicine, University of Puerto Rico, Medical Sciences Campus, PO Box 365067, San Juan, Puerto Rico 00936-5067
| | - José L. Agosto-Rivera
- Department of Biology, University of Puerto Rico, Río Piedras Campus, PO Box 23360, San Juan, Puerto Rico 00931-3360
| | - Amanda J González-Segarra
- Department of Neuroscience and Behavior, Barnard College, Columbia University, New York, New York 10027
| | - Annabell C. Segarra
- Physiology Department, School of Medicine, University of Puerto Rico, Medical Sciences Campus, PO Box 365067, San Juan, Puerto Rico 00936-5067
| |
Collapse
|
19
|
Elaad E, Kochav R, Elkouby T. Lying about money and game points by men and women and its relation to the Self-Reported Lying Scale. Front Psychol 2024; 14:1304237. [PMID: 38298365 PMCID: PMC10827945 DOI: 10.3389/fpsyg.2023.1304237] [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: 10/02/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction The present study was designed to examine the effect of monetary and non-monetary endowment on lying by men and women in the Ultimatum Game. Another goal was to examine to what extent the Self-Reported Lying Scale (SRLS), described here for the first time, predicts lying in the Ultimatum Game. Methods Examinees (162, 82 women) were allocated to four experimental conditions in a 2 × 2 factorial design. Two endowment conditions (money and game points) were crossed with two sex conditions (men and women). Participants underwent an Ultimatum Game in which they were permitted to conceal part of the endowment from an unidentified partner. Finally, participants completed the SRLS. Results The results indicated that more cash than points were concealed from the partner, and men concealed more of their endowment than women. We further defined fake fairness in sharing that combined hiding a more significant portion of the endowment from the partner while presenting fair sharing of the remaining award. We found more fake fairness when money was shared than when points were concealed. Fake fairness is more significant for men than for women. For money and points alike, concealment was predicted by the global score of the SRLS and its five subscales (self-assessed lying ability, lie detection ability, the use of reason in lying, lie acceptability, and lie frequency). Discussion It was suggested that a monetary endowment is more sensitive to lying than game points and involves more fake fairness. Nevertheless, the differences are quantitative, and the same response pattern exists in the two endowment conditions. Replacing money with points is a proper solution whenever a monetary endowment presents difficulties. It was further suggested that sex differences exist in lying using an asymmetric information UG, where proposers were permitted to mislead responders about their endowment. Finally, the SRLS may contribute to a better understanding of the question of who lies.
Collapse
Affiliation(s)
- Eitan Elaad
- Department of Psychology, Ariel University, Ariel, Israel
| | | | | |
Collapse
|
20
|
Iyer V, Saberi SA, Pacheco R, Sizemore EF, Stockman S, Kulkarni A, Cantwell L, Thakur GA, Hohmann AG. Negative allosteric modulation of cannabinoid CB 1 receptor signaling suppresses opioid-mediated tolerance and withdrawal without blocking opioid antinociception. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.06.574477. [PMID: 38260598 PMCID: PMC10802405 DOI: 10.1101/2024.01.06.574477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The direct blockade of CB 1 cannabinoid receptors produces therapeutic effects as well as adverse side-effects that limit their clinical potential. CB 1 negative allosteric modulators (NAMs) represent an indirect approach to decrease the affinity and/or efficacy of orthosteric cannabinoid ligands or endocannabinoids at CB 1 . We recently reported that GAT358, a CB 1 -NAM, blocked opioid-induced mesocorticolimbic dopamine release and reward via a CB 1 -allosteric mechanism of action. Whether a CB 1 -NAM dampens opioid-mediated therapeutic effects such as analgesia or alters other unwanted side-effects of opioids remain unknown. Here, we characterized the effects of GAT358 on nociceptive behaviors in the presence and absence of morphine. We examined the impact of GAT358 on formalin-evoked pain behavior and Fos protein expression, a marker of neuronal activation, in the lumbar dorsal horn. We also assessed the impact of GAT358 on morphine-induced slowing of colonic transit, tolerance, and withdrawal behaviors. GAT358 attenuated morphine antinociceptive tolerance without blocking acute antinociception. GAT358 also reduced morphine-induced slowing of colonic motility without impacting fecal boli production. GAT358 produced antinociception in the presence and absence of morphine in the formalin model of inflammatory nociception and reduced the number of formalin-evoked Fos protein-like immunoreactive cells in the lumbar spinal dorsal horn. Finally, GAT358 mitigated the somatic signs of naloxone-precipitated, but not spontaneous, opioid withdrawal following chronic morphine dosing in mice. Our results support the therapeutic potential of CB 1 -NAMs as novel drug candidates aimed at preserving opioid-mediated analgesia while preventing their unwanted side-effects. Our studies also uncover previously unrecognized antinociceptive properties associated with an arrestin-biased CB 1 -NAMs. Highlights CB 1 negative allosteric modulator (NAM) GAT358 attenuated morphine tolerance GAT358 reduced morphine-induced slowing of colonic motility but not fecal productionGAT358 was antinociceptive for formalin pain alone and when combined with morphineGAT358 reduced formalin-evoked Fos protein expression in the lumbar spinal cordGAT358 mitigated naloxone precipitated withdrawal after chronic morphine dosing.
Collapse
|
21
|
Proaño SB, Miller CK, Krentzel AA, Dorris DM, Meitzen J. Sex steroid hormones, the estrous cycle, and rapid modulation of glutamatergic synapse properties in the striatal brain regions with a focus on 17β-estradiol and the nucleus accumbens. Steroids 2024; 201:109344. [PMID: 37979822 PMCID: PMC10842710 DOI: 10.1016/j.steroids.2023.109344] [Citation(s) in RCA: 1] [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] [Received: 07/25/2023] [Revised: 10/28/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
The striatal brain regions encompassing the nucleus accumbens core (NAcc), shell (NAcs) and caudate-putamen (CPu) regulate cognitive functions including motivated behaviors, habit, learning, and sensorimotor action, among others. Sex steroid hormone sensitivity and sex differences have been documented in all of these functions in both normative and pathological contexts, including anxiety, depression and addiction. The neurotransmitter glutamate has been implicated in regulating these behaviors as well as striatal physiology, and there are likewise documented sex differences in glutamate action upon the striatal output neurons, the medium spiny neurons (MSNs). Here we review the available data regarding the role of steroid sex hormones such as 17β-estradiol (estradiol), progesterone, and testosterone in rapidly modulating MSN glutamatergic synapse properties, presented in the context of the estrous cycle as appropriate. Estradiol action upon glutamatergic synapse properties in female NAcc MSNs is most comprehensively discussed. In the female NAcc, MSNs exhibit development period-specific sex differences and estrous cycle variations in glutamatergic synapse properties as shown by multiple analyses, including that of miniature excitatory postsynaptic currents (mEPSCs). Estrous cycle-differences in NAcc MSN mEPSCs can be mimicked by acute exposure to estradiol or an ERα agonist. The available evidence, or lack thereof, is also discussed concerning estrogen action upon MSN glutamatergic synapse in the other striatal regions as well as the underexplored roles of progesterone and testosterone. We conclude that there is strong evidence regarding estradiol action upon glutamatergic synapse function in female NAcs MSNs and call for more research regarding other hormones and striatal regions.
Collapse
Affiliation(s)
- Stephanie B Proaño
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Christiana K Miller
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Amanda A Krentzel
- Office of Research and Innovation, North Carolina State University, Raleigh, NC, USA
| | - David M Dorris
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- Dept. of Biological Sciences, North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
22
|
Manns A, Torregrossa H, Mahdjoub S, Gomajee R, Melchior M, El-Khoury Lesueur F. Do Determinants of Smoking Cessation and Relapse Differ between Men and Women? Data from a French National Study. Subst Use Misuse 2023; 59:167-176. [PMID: 37813814 DOI: 10.1080/10826084.2023.2267106] [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] [Indexed: 10/11/2023]
Abstract
Introduction: There is limited understanding of different predictors of smoking cessation success (SCS) among women and men, despite well-documented differences in smoking behavior.Methods: Using data from DePICT (Description des Perceptions, Images, et Comportements liés au Tabagisme), a national survey of French adults which recruited 2377 regular and former smokers we investigated whether major determinants of SCS differed by sex. Factors associated with unsuccessful vs. No successful quit attempt; vs. SCS were studied using multivariate multinomial logistic regression analyses stratified by sex.Results: Women and men share some determinants of SCS including no cannabis use, living in a nonsmoker household and importance giving to being a nonsmoker. However, no e-cigarette use, low-to-moderate alcohol consumption, early smoking initiation, and higher education were associated with SCS only among women. No use of nicotine replacement, having family members who smoke, family opinion on smoking and current employment, were associated with SCS only among men. Neutral or negative friends' opinion on smoking or living with a smoker were associated with unsuccessful smoking attempts among men.Conclusions: Our results show differences between determinants of SCS according to sex, which highlights the importance of developing tailored interventions that account for sex/gender differences in smoking cessation.
Collapse
Affiliation(s)
- Aurélia Manns
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| | - Hugo Torregrossa
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| | - Sarah Mahdjoub
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| | - Ramchandar Gomajee
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| | - Maria Melchior
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| | - Fabienne El-Khoury Lesueur
- Department of Social Epidemiology, Sorbonne Université, Institut Pierre Louis d'Epidémiologie et de Santé Publique, INSERM, IPLESP, Paris, France
| |
Collapse
|
23
|
Ethridge SB, Smith MA. Estradiol and Mu opioid-mediated reward: The role of estrogen receptors in opioid use. ADDICTION NEUROSCIENCE 2023; 9:100139. [PMID: 38155959 PMCID: PMC10753849 DOI: 10.1016/j.addicn.2023.100139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2023]
Abstract
Opioid use and opioid use disorder are characterized by sex and gender differences, and some of these differences may be mediated by differences in the hormonal milieu within and across individuals. This review focuses on the role of ovarian hormones, and particularly estradiol, on the endogenous mu opioid receptor system. There is an abundance of data indicating that estradiol influences the activity of endogenous mu opioid peptides, the activation of mu opioid receptors, and the internalization and desensitization of mu opioid receptors. These effects have functional consequences on behaviors mediated by endogenous mu opioid receptor activity and on sensitivity to mu opioid agonists and antagonists. Recent behavioral data suggest these consequences extend to mu opioid reward, and preclinical studies report that estradiol decreases self-administration of mu opioid receptor agonists across a range of experimental conditions. Data collected in human laboratory studies suggest that estradiol may have functionally similar effects in clinical populations, and thus estrogen receptors may be a potential target in the development of novel therapeutics. This review summarizes data from cellular assays to clinical trials to explore how estradiol influences mu opioid receptor activity, as well as potential ways in which estrogen receptors may be targeted to address the problems of opioid use.
Collapse
Affiliation(s)
- Sarah B. Ethridge
- Department of Psychology, Program in Neuroscience, Davidson College, Davidson, NC, USA
| | - Mark A. Smith
- Department of Psychology, Program in Neuroscience, Davidson College, Davidson, NC, USA
| |
Collapse
|
24
|
Estrin DJ, Kulik JM, Beacher NJ, Pawlak AP, Klein SD, West MO. Acquired Alterations in Nucleus Accumbens Responsiveness to a Cocaine-Paired Discriminative Stimulus Preceding Rats' Daily Cocaine Consumption. ADDICTION NEUROSCIENCE 2023; 8:100121. [PMID: 37664217 PMCID: PMC10470667 DOI: 10.1016/j.addicn.2023.100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Resumption of drug taking is a primary focus for substance use disorder research and can be triggered by drug-associated environmental stimuli. The Nucleus Accumbens (NAc) is a key brain region which guides motivated behavior and is implicated in resumption. There remains a pressing need to characterize NAc neurons' responsiveness to drug associated stimuli during withdrawal and abstinence. We recorded discriminative stimulus (DS) induced NAc activity via in vivo single-unit electrophysiology in rats that self-administered cocaine. Male and female rats implanted with a jugular catheter and a microwire array in NAc Core and Shell self-administered cocaine under control of a 30s auditory DS for 6 hours per session across 14 consecutive days. Rats acquired tone discrimination within 4 sessions. To exclude pharmacological effects of circulating cocaine from all neural analyses, we studied changes in DS-induced firing only for trials preceding the first infusion of cocaine in each of the 14 sessions, which were defined as "pre-drug trials." NAc neuron responses were assessed prior to tone-evoked movement onset. Responsiveness to the DS tone was exhibited throughout all sessions by the NAc Core population, but only during Early sessions by the NAc Shell population. Both Core and Shell responded selectively to the DS, i.e., more strongly on drug taking trials, or Hits, than on Missed opportunities. These findings suggest that NAc Core and Shell play distinct roles in initiating cocaine seeking prior to daily cocaine consumption, and align with reports suggesting that as drug use becomes chronic, cue-evoked activity shifts from NAc Shell to NAc Core.
Collapse
Affiliation(s)
- David J. Estrin
- Feil Family Brain & Mind Research Institute, Weill Cornell Medicine, 413 East 69 Street, New York, NY 10021
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| | - Julianna M. Kulik
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| | - Nicholas J. Beacher
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Neural Engineering Section, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD 21224
| | - Anthony P. Pawlak
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Center of Alcohol & Substance Use Studies, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
- Graduate School of Applied & Professional Psychology, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
| | - Samuel D. Klein
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
- Department of Psychology, University of Minnesota-Twin Cities, 75 East River Road, Minneapolis, MN 55455
| | - Mark O. West
- Department of Psychology, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ 08854
| |
Collapse
|
25
|
Santos-Toscano R, Arevalo MA, Garcia-Segura LM, Grassi D, Lagunas N. Interaction of gonadal hormones, dopaminergic system, and epigenetic regulation in the generation of sex differences in substance use disorders: A systematic review. Front Neuroendocrinol 2023; 71:101085. [PMID: 37543184 DOI: 10.1016/j.yfrne.2023.101085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/07/2023]
Abstract
Substance use disorder (SUD) is a chronic condition characterized by pathological drug-taking and seeking behaviors. Remarkably different between males and females, suggesting that drug addiction is a sexually differentiated disorder. The neurobiological bases of sex differences in SUD include sex-specific reward system activation, influenced by interactions between gonadal hormone level changes, dopaminergic reward circuits, and epigenetic modifications of key reward system genes. This systematic review, adhering to PICOS and PRISMA-P 2015 guidelines, highlights the sex-dependent roles of estrogens, progesterone, and testosterone in SUD. In particular, estradiol elevates and progesterone reduces dopaminergic activity in SUD females, whilst testosterone and progesterone augment SUD behavior in males. Finally, SUD is associated with a sex-specific increase in the rate of opioid and monoaminergic gene methylation. The study reveals the need for detailed research on gonadal hormone levels, dopaminergic or reward system activity, and epigenetic landscapes in both sexes for efficient SUD therapy development.
Collapse
Affiliation(s)
- Raquel Santos-Toscano
- School of Medicine, University of Central Lancashire, 135A Adelphi St, Preston PR1 7BH, United Kingdom
| | - Maria Angeles Arevalo
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Miguel Garcia-Segura
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniela Grassi
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Anatomy, Histology and Neuroscience, Autonoma University of Madrid, Calle Arzobispo Morcillo 4, 28029 Madrid, Spain.
| | - Natalia Lagunas
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain; Department of Legal Medicine, Psychiatry and Pathology, School of Medicine, Complutense University of Madrid, Ciudad Universitaria, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| |
Collapse
|
26
|
Petrie J, Kowallis LR, Kamhout S, Bills KB, Adams D, Fleming DE, Brown BL, Steffensen SC. Gender-Specific Interactions in a Visual Object Recognition Task in Persons with Opioid Use Disorder. Biomedicines 2023; 11:2460. [PMID: 37760905 PMCID: PMC10525754 DOI: 10.3390/biomedicines11092460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Opioid use disorder (OUD)-associated overdose deaths have reached epidemic proportions worldwide over the past two decades, with death rates for men reported at twice the rate for women. Using a controlled, cross-sectional, age-matched (18-56 y) design to better understand the cognitive neuroscience of OUD, we evaluated the electroencephalographic (EEG) responses of male and female participants with OUD vs. age- and gender-matched non-OUD controls during a simple visual object recognition Go/No-Go task. Overall, women had significantly slower reaction times (RTs) than men. In addition, EEG N200 and P300 event-related potential (ERP) amplitudes for non-OUD controls were significantly larger for men, while their latencies were significantly shorter than for women. However, while N200 and P300 amplitudes were not significantly affected by OUD for either men or women in this task, latencies were also affected differentially in men vs. women with OUD. Accordingly, for both N200 and P300, male OUD participants exhibited longer latencies while female OUD participants exhibited shorter ones than in non-OUD controls. Additionally, robust oscillations were found in all participants during a feedback message associated with performance in the task. Although alpha and beta power during the feedback message were significantly greater for men than women overall, both alpha and beta oscillations exhibited significantly lower power in all participants with OUD. Taken together, these findings suggest important gender by OUD differences in cognitive processing and reflection of performance in this simple visual task.
Collapse
Affiliation(s)
- JoAnn Petrie
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
| | - Logan R. Kowallis
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
| | - Sarah Kamhout
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
| | - Kyle B. Bills
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
- Department of Neuroscience, Noorda College of Osteopathic Medicine, Provo, UT 84606, USA
| | - Daniel Adams
- PhotoPharmics, Inc., 947 So, 500 E, Suite 100, American Fork, UT 84003, USA
| | - Donovan E. Fleming
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
| | - Bruce L. Brown
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
| | - Scott C. Steffensen
- Department of Psychology, Brigham Young University, Provo, UT 84602, USA; (J.P.); (K.B.B.)
- Department of Neuroscience, Noorda College of Osteopathic Medicine, Provo, UT 84606, USA
| |
Collapse
|
27
|
Knouse MC, Deutschmann AU, Nenov MN, Wimmer ME, Briand LA. Sex differences in pre- and post-synaptic glutamate signaling in the nucleus accumbens core. Biol Sex Differ 2023; 14:52. [PMID: 37596655 PMCID: PMC10439632 DOI: 10.1186/s13293-023-00537-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND Glutamate signaling within the nucleus accumbens underlies motivated behavior and is involved in psychiatric disease. Although behavioral sex differences in these processes are well-established, the neural mechanisms driving these differences are largely unexplored. In these studies, we examine potential sex differences in synaptic plasticity and excitatory transmission within the nucleus accumbens core. Further understanding of baseline sex differences in reward circuitry will shed light on potential mechanisms driving behavioral differences in motivated behavior and psychiatric disease. METHODS Behaviorally naïve adult male and female Long-Evans rats, C57Bl/6J mice, and constitutive PKMζ knockout mice were killed and tissue containing the nucleus accumbens core was collected for ex vivo slice electrophysiology experiments. Electrophysiology recordings examined baseline sex differences in synaptic plasticity and transmission within this region and the potential role of PKMζ in long-term depression. RESULTS Within the nucleus accumbens core, both female mice and rats exhibit higher AMPA/NMDA ratios compared to male animals. Further, female mice have a larger readily releasable pool of glutamate and lower release probability compared to male mice. No significant sex differences were detected in spontaneous excitatory postsynaptic current amplitude or frequency. Finally, the threshold for induction of long-term depression was lower for male animals than females, an effect that appears to be mediated, in part, by PKMζ. CONCLUSIONS We conclude that there are baseline sex differences in synaptic plasticity and excitatory transmission in the nucleus accumbens core. Our data suggest there are sex differences at multiple levels in this region that should be considered in the development of pharmacotherapies to treat psychiatric illnesses such as depression and substance use disorder.
Collapse
Affiliation(s)
- Melissa C Knouse
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Andre U Deutschmann
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Miroslav N Nenov
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Mathieu E Wimmer
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA
| | - Lisa A Briand
- Department of Psychology, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
- Neuroscience Program, Temple University, Weiss Hall, 1701 North 13th Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
28
|
Hersey M, Chen AY, Bartole MK, Anand J, Newman AH, Tanda G. An FSCV Study on the Effects of Targeted Typical and Atypical DAT Inhibition on Dopamine Dynamics in the Nucleus Accumbens Shell of Male and Female Mice. ACS Chem Neurosci 2023; 14:2802-2810. [PMID: 37466616 PMCID: PMC10766117 DOI: 10.1021/acschemneuro.3c00354] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
Understanding the neurochemistry underlying sex differences in psychostimulant use disorders (PSUD) is essential for developing related therapeutics. Many psychostimulants, like cocaine, inhibit the dopamine transporter (DAT), which is largely thought to account for actions related to their misuse and dependence. Cocaine-like, typical DAT inhibitors preferentially bind DAT in an outward-facing conformation, while atypical DAT inhibitors, like modafinil, prefer a more inward-facing DAT conformation. Modafinil and R-modafinil have emerged as potential therapeutic options for selected populations of individuals affected by PSUD. In addition, analogs of modafinil (JJC8-088 and JJC8-091) with different pharmacological profiles have been explored as potential PSUD medications in preclinical models. In this work, we employ fast scan cyclic voltammetry (FSCV) to probe nucleus accumbens shell (NAS) dopamine (DA) dynamics in C57BL/6 male and female mice. We find that cocaine slowed DA clearance in both male and female mice but produced more robust increases in evoked NAS DA in female mice. R-Modafinil produced mild increases in evoked NAS DA and slowed DA clearance across the sexes. The modafinil analog JJC8-088, a typical DAT inhibitor, produced increases in evoked NAS DA in female and male mice. Finally, JJC8-091, an atypical DAT inhibitor, produced limited increases in evoked NAS DA and slowed DA clearance in both sexes. In this work we begin to tease out how sex differences may alter the effects of DAT targeting and highlight how this may help focus research toward effective treatment options for PSUD.
Collapse
Affiliation(s)
- Melinda Hersey
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | - Andy Y. Chen
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | | | - Jayati Anand
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| | - Amy Hauck Newman
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
- Medicinal Chemistry Section, NIDA IRP, Baltimore, MD 21224
| | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224
| |
Collapse
|
29
|
Hersey M, Bartole MK, Jones CS, Newman AH, Tanda G. Are There Prevalent Sex Differences in Psychostimulant Use Disorder? A Focus on the Potential Therapeutic Efficacy of Atypical Dopamine Uptake Inhibitors. Molecules 2023; 28:5270. [PMID: 37446929 PMCID: PMC10343811 DOI: 10.3390/molecules28135270] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Psychostimulant use disorders (PSUD) affect a growing number of men and women and exert sizable public health and economic burdens on our global society. Notably, there are some sex differences in the onset of dependence, relapse rates, and treatment success with PSUD observed in preclinical and clinical studies. The subtle sex differences observed in the behavioral aspects of PSUD may be associated with differences in the neurochemistry of the dopaminergic system between sexes. Preclinically, psychostimulants have been shown to increase synaptic dopamine (DA) levels and may downregulate the dopamine transporter (DAT). This effect is greatest in females during the high estradiol phase of the estrous cycle. Interestingly, women have been shown to be more likely to begin drug use at younger ages and report higher levels of desire to use cocaine than males. Even though there is currently no FDA-approved medication, modafinil, a DAT inhibitor approved for use in the treatment of narcolepsy and sleep disorders, has shown promise in the treatment of PSUD among specific populations of affected individuals. In this review, we highlight the therapeutic potential of modafinil and other atypical DAT inhibitors focusing on the lack of sex differences in the actions of these agents.
Collapse
Affiliation(s)
| | | | | | | | - Gianluigi Tanda
- Medication Development Program, NIDA IRP, Baltimore, MD 21224, USA; (M.H.); (M.K.B.); (C.S.J.); (A.H.N.)
| |
Collapse
|
30
|
Gao W, Pan T, Fan G, Cui J, Wang T, Huang N, Jiang C, Ma L, Wang F, Liu X, Le Q. Enhanced heroin analgesic effect in male offspring of sires who self-administered heroin. Front Pharmacol 2023; 14:1211897. [PMID: 37388448 PMCID: PMC10303812 DOI: 10.3389/fphar.2023.1211897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/30/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction: A growing body of evidence suggests that parental substance abuse, even prior to conception, may induce phenotypic changes in offspring. Parental opioid exposure has been shown to affect developmental processes, induce memory deficits, and lead to psycho-emotional disorders in offspring. However, how parental, especially paternal, chronic drug exposure affects offspring remains unexplored. Methods: Adult male rats were subjected to 31 days of heroin self-administration followed by mating with naïve females. Litter size and body weight of F1 offspring were recorded. Object-based attention tests, cocaine self-administration tests, and hot plate tests were used to test for potential effects of chronic paternal heroin seeking on cognition, reward, or analgesic sensitivity in the offspring. Results: Body weight and litter size of the heroin F1 generation were not altered compared to the saline F1 generation. Furthermore, paternal chronic heroin self-administration experience had no significant effect on object-based attention tests or cocaine self-administration behavior in either sex. However, in the hot plate test, although no difference in basal latency was found between the two groups in either sex, a significant increase in the analgesic effect of heroin was observed in the male heroin F1 generation. Conclusions: Taken together, these data provide evidence that paternal chronic heroin self-administration experience could sex-dimorphically increase the analgesic effect of heroin in male offspring, but had no significant effect on response to cocaine reinforcement or attentional behavior.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lan Ma
- *Correspondence: Qiumin Le, ; Lan Ma,
| | | | | | - Qiumin Le
- *Correspondence: Qiumin Le, ; Lan Ma,
| |
Collapse
|
31
|
Corbett CM, Miller EN, Wannen EE, Rood BD, Chandler DJ, Loweth JA. Cocaine Exposure Increases Excitatory Synaptic Transmission and Intrinsic Excitability in the Basolateral Amygdala in Male and Female Rats and across the Estrous Cycle. Neuroendocrinology 2023; 113:1127-1139. [PMID: 37271140 PMCID: PMC10623393 DOI: 10.1159/000531351] [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: 01/23/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Sex and ovarian hormones influence cocaine seeking and relapse vulnerability, but less is known regarding the cellular and synaptic mechanisms contributing to these behavioral sex differences. One factor thought to influence cue-induced seeking behavior following withdrawal is cocaine-induced changes in the spontaneous activity of pyramidal neurons in the basolateral amygdala (BLA). However, the mechanisms underlying these changes, including potential sex or estrous cycle effects, are unknown. METHODS Ex vivo whole-cell patch clamp electrophysiology was conducted to investigate the effects of cocaine exposure, sex, and estrous cycle fluctuations on two properties that can influence spontaneous activity of BLA pyramidal neurons: (1) frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) and (2) intrinsic excitability. Recordings of BLA pyramidal neurons were conducted in adult male and female rats and across the estrous cycle following 2-4 weeks of withdrawal from extended-access cocaine self-administration (6 h/day for 10 days) or drug-naïve conditions. RESULTS In both sexes, cocaine exposure increased the frequency, but not amplitude, of sEPSCs and neuronal intrinsic excitability. Across the estrous cycle, sEPSC frequency and intrinsic excitability were significantly elevated only in cocaine-exposed females in the estrus stage of the cycle, a stage when cocaine-seeking behavior is known to be enhanced. CONCLUSIONS Here, we identify potential mechanisms underlying cocaine-induced alterations in the spontaneous activity of BLA pyramidal neurons in both sexes along with changes in these properties across the estrous cycle.
Collapse
Affiliation(s)
- Claire M. Corbett
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Emily N.D. Miller
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Erin E. Wannen
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Benjamin D Rood
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Daniel J. Chandler
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| | - Jessica A. Loweth
- Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, New Jersey, USA
| |
Collapse
|
32
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. Mol Cell Neurosci 2023; 125:103849. [PMID: 36965548 PMCID: PMC10898607 DOI: 10.1016/j.mcn.2023.103849] [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: 01/13/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 03/27/2023] Open
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
33
|
Krueger K, Lamenza F, Gu H, El-Hodiri H, Wester J, Oberdick J, Fischer AJ, Oghumu S. Sex differences in susceptibility to substance use disorder: Role for X chromosome inactivation and escape? Mol Cell Neurosci 2023; 125:103859. [PMID: 37207894 PMCID: PMC10286730 DOI: 10.1016/j.mcn.2023.103859] [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: 12/26/2022] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/21/2023] Open
Abstract
There is a sex-based disparity associated with substance use disorders (SUDs) as demonstrated by clinical and preclinical studies. Females are known to escalate from initial drug use to compulsive drug-taking behavior (telescoping) more rapidly, and experience greater negative withdrawal effects than males. Although these biological differences have largely been attributed to sex hormones, there is evidence for non-hormonal factors, such as the influence of the sex chromosome, which underlie sex disparities in addiction behavior. However, genetic and epigenetic mechanisms underlying sex chromosome influences on substance abuse behavior are not completely understood. In this review, we discuss the role that escape from X-chromosome inactivation (XCI) in females plays in sex-associated differences in addiction behavior. Females have two X chromosomes (XX), and during XCI, one X chromosome is randomly chosen to be transcriptionally silenced. However, some X-linked genes escape XCI and display biallelic gene expression. We generated a mouse model using an X-linked gene specific bicistronic dual reporter mouse as a tool to visualize allelic usage and measure XCI escape in a cell specific manner. Our results revealed a previously undiscovered X-linked gene XCI escaper (CXCR3), which is variable and cell type dependent. This illustrates the highly complex and context dependent nature of XCI escape which is largely understudied in the context of SUD. Novel approaches such as single cell RNA sequencing will provide a global molecular landscape and impact of XCI escape in addiction and facilitate our understanding of the contribution of XCI escape to sex disparities in SUD.
Collapse
Affiliation(s)
- Kate Krueger
- Department of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Felipe Lamenza
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Microbiology, The Ohio State University, Columbus, OH, USA
| | - Howard Gu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | - Heithem El-Hodiri
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Jason Wester
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - John Oberdick
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Steve Oghumu
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
34
|
Chapp AD, Nwakama CA, Thomas MJ, Meisel RL, Mermelstein PG. Sex Differences in Cocaine Sensitization Vary by Mouse Strain. Neuroendocrinology 2023; 113:1167-1176. [PMID: 37040721 DOI: 10.1159/000530591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/30/2023] [Indexed: 04/13/2023]
Abstract
INTRODUCTION Preclinical literature, frequently utilizing rats, suggests females display a more rapid advancement of substance abuse and a greater risk of relapse following drug abstinence. In clinical populations, it is less clear as to what extent biological sex is a defining variable in the acquisition and maintenance of substance use. Even without considering environmental experiences, genetic factors are presumed to critically influence the vulnerability to addiction. Genetically diverse mouse models provide a robust tool to examine the interactions between genetic background and sex differences in substance abuse. METHODS We explored mouse strain variability in male versus female behavioral sensitization to cocaine. Locomotor sensitization was observed following 5 consecutive days of subcutaneous cocaine across three genetically different mice strains: C57BL/6J, B6129SF2/J, and Diversity Outbred (DO/J). RESULTS Sex differences in cocaine locomotor sensitization were dependent on mouse strain. Specifically, we observed opposing sex differences in locomotor sensitization, with male C57BL/6J and female B6129SF2/J mice displaying heightened activity compared to their opposite sex counterparts. Conversely, no sex differences were observed in the DO/J mice. Acute cocaine administration resulted in locomotor differences across strains in male, but not female, mice. The magnitude of sensitization (or lack thereof) also varied by genetic background. CONCLUSIONS While sex differences in drug addiction may be observed, these effects can be mitigated, or even reversed, depending on genetic background. The clinical implications are that in the absence of understanding the genetic variables underlying vulnerability to addiction, sex provides little information regarding the predisposition of an individual to drug abuse.
Collapse
Affiliation(s)
- Andrew D Chapp
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Chinonso A Nwakama
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark J Thomas
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Robert L Meisel
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul G Mermelstein
- Department of Neuroscience and Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
35
|
Maher EE, Strzelecki AM, Weafer JJ, Gipson CD. The importance of translationally evaluating steroid hormone contributions to substance use. Front Neuroendocrinol 2023; 69:101059. [PMID: 36758769 PMCID: PMC10182261 DOI: 10.1016/j.yfrne.2023.101059] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023]
Abstract
Clinically, women appear to be more susceptible to certain aspects of substance use disorders (SUDs). The steroid hormones 17β-estradiol (E2) and progesterone (Pg) have been linked to women-specific drug behaviors. Here, we review clinical and preclinical studies investigating how cycling ovarian hormones affect nicotine-, cocaine-, and opioid-related behaviors. We also highlight gaps in the literature regarding how synthetic steroid hormone use may influence drug-related behaviors. In addition, we explore how E2 and Pg are known to interact in brain reward pathways and provide evidence of how these interactions may influence drug-related behaviors. The synthesis of this review demonstrates the critical need to study women-specific factors that may influence aspects of SUDs, which may play important roles in addiction processes in a sex-specific fashion. It is important to understand factors that impact women's health and may be key to moving the field forward toward more efficacious and individualized treatment strategies.
Collapse
Affiliation(s)
- Erin E Maher
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States
| | - Ashley M Strzelecki
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Jessica J Weafer
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Cassandra D Gipson
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
36
|
Lamanna-Rama N, MacDowell KS, López G, Leza JC, Desco M, Ambrosio E, Soto-Montenegro ML. Neuroimaging revealed long-lasting glucose metabolism changes to morphine withdrawal in rats pretreated with the cannabinoid agonist CP-55,940 during periadolescence. Eur Neuropsychopharmacol 2023; 69:60-76. [PMID: 36780817 DOI: 10.1016/j.euroneuro.2023.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/28/2022] [Accepted: 01/22/2023] [Indexed: 02/13/2023]
Abstract
This study evaluates the long-term effects of a six and 14-week morphine withdrawal in rats pretreated with a cannabinoid agonist (CP-55,940, CP) during periadolescence. Wistar rats (33 males; 32 females) were treated with CP or its vehicle (VH) from postnatal day (PND) 28-38. At PND100, rats performed morphine self-administration (MSA, 15d/12 h/session). Eight groups were defined according to pretreatment (CP), treatment (morphine), and sex. Three [18F]FDG-PET brain images were acquired: after MSA, and after six and 14 weeks of withdrawal. PET data were analyzed with SPM12. Endocannabinoid (EC) markers were evaluated in frozen brain tissue at endpoint. Females showed a higher mean number of self-injections than males. A main Sex effect on global brain metabolism was found. FDG uptake in males was discrete, whereas females showed greater brain metabolism changes mainly in areas of the limbic system after morphine treatment. Moreover, the morphine-induced metabolic pattern in females was exacerbated when CP was previously present. In addition, the CP-Saline male group showed reduced CB1R, MAGL expression, and NAPE/FAAH ratio compared to the control group, and morphine was able to reverse CB1R and MAGL expression almost to control levels. In conclusion, females showed greater and longer-lasting metabolic changes after morphine withdrawal than males, indicating a higher vulnerability and a different sensitivity to morphine in subjects pre-exposed to CP. In contrast, males primarily showed changes in EC markers. Together, our results suggest that CP pre-exposure contributes to the modulation of brain metabolism and EC systems in a sex-dependent manner.
Collapse
Affiliation(s)
- N Lamanna-Rama
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain
| | - K S MacDowell
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - G López
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain; Faculty of Health Science, Universidad Internacional de La Rioja (UNIR), Spain
| | - J C Leza
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Departamento de Farmacología & Toxicología, Facultad de Medicina, Universidad Complutense de Madrid, Imas12, IUIN, Spain
| | - M Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Leganés, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | - E Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, National University for Distance Learning (UNED), Madrid, Spain.
| | - M L Soto-Montenegro
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain; CIBER de Salud Mental (CIBERSAM), Madrid, Spain; Grupo de Fisiopatología y Farmacología del Sistema Digestivo de la Universidad Rey Juan Carlos (NEUGUT), Madrid, España.
| |
Collapse
|
37
|
Miller CK, Krentzel AA, Meitzen J. ERα Stimulation Rapidly Modulates Excitatory Synapse Properties in Female Rat Nucleus Accumbens Core. Neuroendocrinology 2023; 113:1140-1153. [PMID: 36746131 PMCID: PMC10623399 DOI: 10.1159/000529571] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/30/2023] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The nucleus accumbens core (NAcc) is a sexually differentiated brain region that is modulated by steroid hormones such as 17β-estradiol (estradiol), with consequential impacts on relevant motivated behaviors and disorders such as addiction, anxiety, and depression. NAcc estradiol levels naturally fluctuate, including during the estrous cycle in adult female rats, which is analogous to the menstrual cycle in adult humans. Across the estrous cycle, excitatory synapse properties of medium spiny neurons rapidly change, as indicated by analysis of miniature excitatory postsynaptic currents (mEPSCs). mEPSC frequency decreases during estrous cycle phases associated with high estradiol levels. This decrease in mEPSC frequency is mimicked by acute topical exposure to estradiol. The identity of the estrogen receptor (ER) underlying this estradiol action is unknown. Adult rat NAcc expresses three ERs, all extranuclear: membrane ERα, membrane ERβ, and GPER1. METHODS In this brief report, we take a first step toward addressing this challenge by testing whether activation of ERs via acute topical agonist application is sufficient for inducing changes in mEPSC properties recorded via whole-cell patch clamp. RESULTS An agonist of ERα induced large decreases in mEPSC frequency, while agonists of ERβ and GPER1 did not robustly modulate mEPSC properties. CONCLUSIONS These data provide evidence that activation of ERα is sufficient for inducing changes in mEPSC frequency and is a likely candidate underlying the estradiol-induced changes observed during the estrous cycle. Overall, these findings extend our understanding of the neuroendocrinology of the NAcc and implicate ERα as a primary target for future studies.
Collapse
Affiliation(s)
- Christiana K. Miller
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Amanda A. Krentzel
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
38
|
Phillips RA, Tuscher JJ, Fitzgerald ND, Wan E, Zipperly ME, Duke CG, Ianov L, Day JJ. Distinct subpopulations of D1 medium spiny neurons exhibit unique transcriptional responsiveness to cocaine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523845. [PMID: 36711527 PMCID: PMC9882178 DOI: 10.1101/2023.01.12.523845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Drugs of abuse increase extracellular concentrations of dopamine in the nucleus accumbens (NAc), resulting in transcriptional alterations that drive long-lasting cellular and behavioral adaptations. While decades of research have focused on the transcriptional mechanisms by which drugs of abuse influence neuronal physiology and function, few studies have comprehensively defined NAc cell type heterogeneity in transcriptional responses to drugs of abuse. Here, we used single nucleus RNA-seq (snRNA-seq) to characterize the transcriptome of over 39,000 NAc cells from male and female adult Sprague-Dawley rats following acute or repeated cocaine experience. This dataset identified 16 transcriptionally distinct cell populations, including two populations of medium spiny neurons (MSNs) that express the Drd1 dopamine receptor (D1-MSNs). Critically, while both populations expressed classic marker genes of D1-MSNs, only one population exhibited a robust transcriptional response to cocaine. Validation of population-selective transcripts using RNA in situ hybridization revealed distinct spatial compartmentalization of these D1-MSN populations within the NAc. Finally, analysis of published NAc snRNA-seq datasets from non-human primates and humans demonstrated conservation of MSN subtypes across rat and higher order mammals, and further highlighted cell type-specific transcriptional differences across the NAc and broader striatum. These results highlight the utility in using snRNA-seq to characterize both cell type heterogeneity and cell type-specific responses to cocaine and provides a useful resource for cross-species comparisons of NAc cell composition.
Collapse
Affiliation(s)
- Robert A. Phillips
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer J. Tuscher
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - N. Dalton Fitzgerald
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ethan Wan
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Morgan E. Zipperly
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corey G. Duke
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lara Ianov
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jeremy J. Day
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
39
|
Fox HC, Milivojevic V, Sinha R. Therapeutics for Substance-Using Women: The Need to Elucidate Sex-Specific Targets for Better-Tailored Treatments. Handb Exp Pharmacol 2023; 282:127-161. [PMID: 37592081 DOI: 10.1007/164_2023_687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
In the last decade, alcohol consumption in the US has risen by 84% in women compared with 35% in men. Furthermore, research has shown that sex- and gender-related differences may disadvantage women in terms of developing a range of psychological, cognitive, and medical problems considerably earlier in their drinking history than men, and despite consuming a similar quantity of substances. While this "telescoping" process has been acknowledged in the literature, a concomitant understanding of the underlying biobehavioral mechanisms, and an increase in the development of specific treatments tailored to women, has not occurred. In the current chapter we focus on understanding why the need for personalized, sex-specific medications is imperative, and highlight some of the potential sex-specific gonadal and stress-related adaptations underpinning the accelerated progress from controlled to compulsive drug and alcohol seeking in women. We additionally discuss the efficacy of these mechanisms as novel targets for medications development, using exogenous progesterone and guanfacine as examples. Finally, we assess some of the challenges faced and progress made in terms of developing innovative medications in women. We suggest that agents such as exogenous progesterone and adrenergic medications, such as guanfacine, may provide some efficacy in terms of attenuating stress-induced craving for several substances, as well as improving the ability to emotionally regulate in the face of stress, preferentially in women. However, to fully leverage the potential of these therapeutics in substance-using women, greater focus needs to the placed on reducing barriers to treatment and research by encouraging women into clinical trials.
Collapse
Affiliation(s)
- Helen C Fox
- Department of Psychiatry and Behavioral Health, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | - Verica Milivojevic
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- The Yale Stress Center, Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
40
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
41
|
DeVito EE, Sofuoglu M. Catechol-O-Methyltransferase Effects on Smoking: A Review and Proof of Concept of Sex-Sensitive Effects. Curr Behav Neurosci Rep 2022; 9:113-123. [PMID: 36644316 PMCID: PMC9838826 DOI: 10.1007/s40473-022-00251-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/19/2023]
Abstract
Purpose of Review This article reviews recent research on how catechol-O-methyltransferase (COMT) may impact cigarette smoking behavior, and how effects may be sex-sensitive. Preliminary data are presented on sex-sensitive effects of COMT on response to short-term abstinence in individuals who smoke. Recent Findings Although research is mixed, functional variants in the COMT gene have been linked with smoking behavior, cessation outcomes and nicotine abstinence-related symptoms. Our proof-of-concept preliminary data from a human laboratory study of individuals who smoke cigarettes found that those with the high COMT enzyme activity genotype (Val/Val) reported more severe smoking urges and withdrawal symptoms following overnight abstinence than Met carriers. These effects were present in women, but not in men and were abstinent-dependent, in that they dissipated following nicotine administration. Summary The preliminary data showing sex-sensitive pharmacogenetic effects may shed light on mechanisms contributing to sex differences in barriers to smoking cessation or potential sex-specific treatment options.
Collapse
Affiliation(s)
- Elise E. DeVito
- Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA
| | - Mehmet Sofuoglu
- Yale University School of Medicine, Department of Psychiatry, New Haven, CT, USA
- VA Connecticut Healthcare System, West Haven, CT
| |
Collapse
|
42
|
Iyer V, Rangel-Barajas C, Woodward TJ, Kulkarni A, Cantwell L, Crystal JD, Mackie K, Rebec GV, Thakur GA, Hohmann AG. Negative allosteric modulation of CB 1 cannabinoid receptor signaling suppresses opioid-mediated reward. Pharmacol Res 2022; 185:106474. [PMID: 36179954 PMCID: PMC9948526 DOI: 10.1016/j.phrs.2022.106474] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/15/2022] [Accepted: 09/25/2022] [Indexed: 01/18/2023]
Abstract
Blockade of cannabinoid type 1 (CB1)-receptor signaling decreases the rewarding properties of many drugs of abuse and has been proposed as an anti-addiction strategy. However, psychiatric side-effects limit the clinical potential of orthosteric CB1 antagonists. Negative allosteric modulators (NAMs) represent a novel and indirect approach to attenuate CB1 signaling by decreasing affinity and/or efficacy of CB1 ligands. We hypothesized that a CB1-NAM would block opioid reward while avoiding the unwanted effects of orthosteric CB1 antagonists. GAT358, a CB1-NAM, failed to elicit cardinal signs of direct CB1 activation or inactivation when administered by itself. GAT358 decreased catalepsy and hypothermia but not antinociception produced by the orthosteric CB1 agonist CP55,940, suggesting that a CB1-NAM blocked cardinal signs of CB1 activation. Next, GAT358 was evaluated using in vivo assays of opioid-induced dopamine release and reward in male rodents. In the nucleus accumbens shell, a key component of the mesocorticolimbic reward pathway, morphine increased electrically-evoked dopamine efflux and this effect was blocked by a dose of GAT358 that lacked intrinsic effects on evoked dopamine efflux. Moreover, GAT358 blocked morphine-induced reward in a conditioned place preference (CPP) assay without producing reward or aversion alone. GAT358-induced blockade of morphine CPP was also occluded by GAT229, a CB1 positive allosteric modulator (CB1-PAM), and absent in CB1-knockout mice. Finally, GAT358 also reduced oral oxycodone (but not water) consumption in a two-bottle choice paradigm. Our results support the therapeutic potential of CB1-NAMs as novel drug candidates aimed at preventing opioid reward and treating opioid abuse while avoiding unwanted side-effects.
Collapse
Affiliation(s)
- Vishakh Iyer
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | | | - Taylor J. Woodward
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Lucas Cantwell
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Jonathon D. Crystal
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ken Mackie
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA
| | - George V. Rebec
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Andrea G. Hohmann
- Program in Neuroscience, Indiana University, Bloomington, IN, USA,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA,Gill Center for Biomolecular Science, Indiana University, Bloomington, IN, USA,Corresponding Author: Andrea G. Hohmann, Psychological and Brain Sciences, Gill Center for Biomolecular Science, Indiana University, Bloomington, IN 47405-7007,
| |
Collapse
|
43
|
Wang L, Zheng H, Wang M, Chen S, Du X, Dong GH. Sex differences in neural substrates of risk taking: Implications for sex-specific vulnerabilities to internet gaming disorder. J Behav Addict 2022; 11:778-795. [PMID: 36053718 PMCID: PMC9872542 DOI: 10.1556/2006.2022.00057] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 06/15/2022] [Accepted: 07/20/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND AND AIMS Sex differences in internet gaming disorder (IGD) remain unknown. Investigating sex-specific neural features that underlie the core risk factor (i.e., risk-taking) of IGD would help in understanding sex-specific vulnerabilities to IGD and advance sex-specific treatments and prevention for IGD. METHODS 111 participants (28 IGD males, 27 IGD females, 26 recreational game user (RGU) males, 30 RGU females) completed a probability discounting task during fMRI scanning. RESULTS First, among RGUs, males showed a higher risk-taking tendency and greater neural activation associated with risk/value evaluation for reward (the ventromedial prefrontal cortex (vmPFC), anterior cingulate cortex (ACC), left putamen) and smaller activation associated with cognitive control (the inferior frontal gyrus) than females during the contrast of risky-safe choices. Moreover, males showed a greater modulatory effect of risky choices on the connection from the vmPFC/ACC to the left putamen than females. Second, IGD males showed decreased activation in the vmPFC/ACC and left putamen compared to RGU males, whereas this decrease did not exist in IGD females. DISCUSSION Males show a higher risk-taking tendency than females. Altered neural substrates associated with risky decision-making exist in IGD males but not in IGD females. CONCLUSIONS The present findings fill the gap in information on the behavioral and neural substrates underlying IGD among females and demonstrate that a high risk-taking tendency is a risk factor and core symptom only in IGD males but not in IGD females. It is necessary to design and adopt distinct treatments and prevention strategies for IGD in males and females.
Collapse
Affiliation(s)
- Lingxiao Wang
- Centre for Cognition and Brain disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Institute of Psychological Science, Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang Province, China,Corresponding author. E-mail:
| | - Hui Zheng
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Wang
- Centre for Cognition and Brain disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Institute of Psychological Science, Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang Province, China
| | - Shuaiyu Chen
- Centre for Cognition and Brain disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Institute of Psychological Science, Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang Province, China
| | - Xiaoxia Du
- School of Psychology, Shanghai University of Sport, Shanghai, China
| | - Guang-Heng Dong
- Centre for Cognition and Brain disorders, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Institute of Psychological Science, Hangzhou Normal University, Hangzhou, Zhejiang Province, China,Zhejiang Key Laboratory for Research in Assessment of Cognitive Impairments, Hangzhou, Zhejiang Province, China,Corresponding author. E-mail:
| |
Collapse
|
44
|
Albores-Garcia D, Stansfield KH, McGlothan JL, Bursac Z, Guilarte TR. Chronic early-life lead exposure sensitizes adolescent rats to cocaine: Role of the dopaminergic system. Front Mol Neurosci 2022; 15:946726. [PMID: 36090247 PMCID: PMC9450041 DOI: 10.3389/fnmol.2022.946726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Exposure to heavy metals has been associated with psychiatric disorders and recent studies suggest an association between childhood lead (Pb2+) intoxication and schizophrenia (SZ). In animal models, Pb2+ exposure recapitulates key neuropathological and dopaminergic system alterations present in SZ. Given the high comorbidity of mental disorders such as SZ and substance abuse, coupled with evidence showing that Pb2+ exposure affects addiction circuits, we hypothesized that early life Pb2+ exposure could sensitize neuronal systems relevant to SZ and substance abuse. To this goal, we examined the effects of chronic developmental Pb2+ exposure on the acute locomotor response to cocaine (0, 5, and 15 mg kg–1) and behavioral sensitization. We also examined the role of the dopaminergic system in the psychostimulant effects of cocaine, and measured D1-dopamine receptor (D1R) levels in the rat brain using [3H]-SCH23390 quantitative receptor autoradiography, as well as the ability of the D1R antagonist SCH23390 to block the cocaine effects on locomotor activation. These studies were performed in male and female rats at different developmental ages consisting of juveniles (postnatal, PN14), early-adolescent (PN28), late adolescent (PN50), and adults (PN120). Our results show that chronic developmental Pb2+ exposure increases the acute locomotor response to the higher dose of cocaine in Pb2+-exposed male adolescent (PN28 and PN50) rats, and to the lower dose of cocaine in adolescent female rats. No changes in the locomotor activity were detected in adult rats. Behavioral sensitization experiments showed a sustained sensitization in early adolescent Pb2+-exposed male but not female rats. The cocaine-induced effects on locomotor activity were abrogated by injection of a D1R antagonist suggesting the involvement of this dopamine receptor subtype. Furthermore, Pb2+-induced increases D1R levels in several brain regions were prominent in juveniles and early adolescence but not in late adolescence or in adults. In summary, early chronic developmental Pb2+ exposure results in age and sex-dependent effect on the locomotor response to cocaine, suggesting differential susceptibilities to the neurotoxic effects of Pb2+ exposure. Our data provides further support to the notion that Pb2+ exposure is an environmental risk factor for psychiatric disorders and substance abuse.
Collapse
Affiliation(s)
- Damaris Albores-Garcia
- Brain, Behavior and the Environment Laboratory, Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | | | - Jennifer L. McGlothan
- Brain, Behavior and the Environment Laboratory, Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Zoran Bursac
- Department of Biostatistics, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
| | - Tomás R. Guilarte
- Brain, Behavior and the Environment Laboratory, Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, United States
- *Correspondence: Tomás R. Guilarte,
| |
Collapse
|
45
|
Krentzel AA, Proaño SB, Dorris DM, Setzer B, Meitzen J. The estrous cycle and 17β-estradiol modulate the electrophysiological properties of rat nucleus accumbens core medium spiny neurons. J Neuroendocrinol 2022; 34:e13122. [PMID: 35365910 PMCID: PMC9250601 DOI: 10.1111/jne.13122] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 12/03/2022]
Abstract
The nucleus accumbens core is a key nexus within the mammalian brain for integrating the premotor and limbic systems and regulating important cognitive functions such as motivated behaviors. Nucleus accumbens core functions show sex differences and are sensitive to the presence of hormones such as 17β-estradiol (estradiol) in normal and pathological contexts. The primary neuron type of the nucleus accumbens core, the medium spiny neuron (MSN), exhibits sex differences in both intrinsic excitability and glutamatergic excitatory synapse electrophysiological properties. Here, we provide a review of recent literature showing how estradiol modulates rat nucleus accumbens core MSN electrophysiology within the context of the estrous cycle. We review the changes in MSN electrophysiological properties across the estrous cycle and how these changes can be mimicked in response to exogenous estradiol exposure. We discuss in detail recent findings regarding how acute estradiol exposure rapidly modulates excitatory synapse properties in nucleus accumbens core but not caudate-putamen MSNs, which mirror the natural changes seen across estrous cycle phases. These recent insights demonstrate the strong impact of sex-specific estradiol action upon nucleus accumbens core neuron electrophysiology.
Collapse
Affiliation(s)
- Amanda A. Krentzel
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Stephanie B. Proaño
- Neurobiology LaboratoryNational Institute of Environmental Health Sciences, NIHResearch Triangle ParkNCUSA
| | - David M. Dorris
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
| | - Beverly Setzer
- Graduate Program for Neuroscience and Department of Biomedical EngineeringBoston UniversityBostonMAUSA
| | - John Meitzen
- Department of Biological SciencesNorth Carolina State UniversityRaleighNCUSA
- Comparative Medicine InstituteNorth Carolina State UniversityRaleighNCUSA
- Center for Human Health and the EnvironmentNorth Carolina State UniversityRaleighNCUSA
| |
Collapse
|
46
|
López-Toro E, Wolf CJH, González RA, van den Brink W, Schellekens A, Vélez-Pastrana MC. Network Analysis of DSM Symptoms of Substance Use Disorders and Frequently Co-Occurring Mental Disorders in Patients with Substance Use Disorder Who Seek Treatment. J Clin Med 2022; 11:jcm11102883. [PMID: 35629008 PMCID: PMC9145186 DOI: 10.3390/jcm11102883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/10/2022] [Accepted: 05/15/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Substance use disorders (SUD) often co-occur with other psychiatric conditions. Research on SUD and comorbid disorders generally flows from a categorical diagnostic or dimensional latent variable perspective, where symptoms are viewed as independent indicators of an underlying disorder. In contrast, the current study took a network analysis perspective to examine the relationships between DSM symptoms of SUD, ADHD, conduct disorder (CD), depression (MDD), and borderline personality disorder (BPD). In addition, we explored possible gender differences in the network structures of these symptoms. Method: In a sample of 722 adult treatment-seeking patients with SUD from the International ADHD in Substance Use Disorders Prevalence Study (IASP) we estimated the network structure for 41 symptoms of SUD, ADHD, CD, MDD, and BPD. We described the structure of symptom networks and their characteristics for the total sample, and we compared the symptom networks for males and females. Results: Network analyses identified seven clusters of symptoms, largely corresponding with the DSM diagnostic categories. There were some connections between clusters, mainly between some hyperactivity symptoms and CD and depressive symptoms. ADHD hyperactivity was most central in the symptom network. Invariance tests revealed no significant gender differences in the structure of symptom networks. Conclusions: The current findings support the categorical DSM classification of mental disorders in treatment-seeking patients with SUD. Future network analyses should include a broader range of symptoms and prospectively explore changes in the symptoms network of patients during treatment.
Collapse
Affiliation(s)
- Edith López-Toro
- PhD Program in Clinical Psychology, Universidad Carlos Albizu, P.O. Box 9023711, San Juan, PR 00902-3711, USA;
| | - Casper J. H. Wolf
- Department of Psychiatry, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (C.J.H.W.); (A.S.)
- Department of Cognitive Neuroscience, Donders Institute for Brain Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, The Netherlands
- Nijmegen Institute for Scientist-Practitioners in Addiction (NISPA), 6525 HR Nijmegen, The Netherlands
| | - Rafael A. González
- National Adoption and Fostering Service & National Conduct Problems Team, Michael Rutter Centre, South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK;
- Centre for Psychiatry, Imperial College London, London W12 0NN, UK
| | - Wim van den Brink
- Department of Psychiatry, Amsterdam University Medical Center, Location Academic Medical Center, Meibergdreef 5, 1105 AZ Amsterdam, The Netherlands;
| | - Arnt Schellekens
- Department of Psychiatry, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (C.J.H.W.); (A.S.)
- Nijmegen Institute for Scientist-Practitioners in Addiction (NISPA), 6525 HR Nijmegen, The Netherlands
| | - María C. Vélez-Pastrana
- PhD Program in Clinical Psychology, Universidad Carlos Albizu, P.O. Box 9023711, San Juan, PR 00902-3711, USA;
- Correspondence: ; Tel.: +1-787-725-6500
| | | |
Collapse
|
47
|
Dolgetta A, Johnson M, Fruitman K, Siegel L, Zhou Y, McEwen BS, Kreek MJ, Milner TA. Sex and chronic stress alter the distribution of glutamate receptors within rat hippocampal CA3 pyramidal cells following oxycodone conditioned place preference. Neurobiol Stress 2022; 17:100431. [PMID: 35535260 PMCID: PMC9076964 DOI: 10.1016/j.ynstr.2022.100431] [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] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/02/2022] [Accepted: 01/19/2022] [Indexed: 12/01/2022] Open
Abstract
Glutamate receptors have a key role in the neurobiology of opioid addiction. Using electron microscopic immunocytochemical methods, this project elucidates how sex and chronic immobilization stress (CIS) impact the redistribution of GluN1 and GluA1 within rat hippocampal CA3 pyramidal cells following oxycodone (Oxy) conditioned place preference (CPP). Four groups of female and male Sprague-Dawley rats subjected to CPP were used: Saline- (Sal) and Oxy-injected (3 mg/kg, I.P.) naïve rats; and Sal- and Oxy-injected CIS rats. GluN1: In both naive and CIS rats, Sal-females compared to Sal-males had elevated cytoplasmic and total dendritic GluN1. Following Oxy CPP, near plasmalemmal, cytoplasmic, and total GluN1 decreased in CA3 dendrites of unstressed females suggesting reduced pools of GluN1 available for ligand binding. Following CIS, Oxy-males (which did not acquire CPP) had increased GluN1 in all compartments of dendrites and spines of CA3 neurons. GluA1: There were no differences in the distribution GluA1 in any cellular compartments of CA3 dendrites in naïve females and males following either Sal or Oxy CPP. CIS alone increased the percent of GluA1 in CA3 dendritic spines in males compared to females. CIS Oxy-males compared to CIS Sal-males had an increase in cytoplasmic and total dendritic GluA1. Thus, in CIS Oxy-males increased pools of GluN1 and GluA1 are available for ligand binding in CA3 neurons. Together with our prior experiments, these changes in GluN1 and GluA1 following CIS in males may contribute to an increased sensitivity of CA3 neurons to glutamate excitation and a reduced capacity to acquire Oxy CPP.
Collapse
Key Words
- ABC, avidin-biotin complex
- AMPA receptors
- BSA, bovine serum albumin
- CIS, chronic immobilization stress
- CPP, conditioned place preference
- DAB, diaminobenzidine
- DG, dentate gyrus
- DOR, delta opioid receptor
- Drug associative-learning
- Electron microscopy
- GABA, Gamma-amino butyric acid
- GluA1, AMPA glutamate receptor subunit 1
- GluN1, NMDA, glutamate receptor subunit 1
- LTP, long-term potentiation
- MOR, mu opioid receptor
- NMDA receptors
- NMDA, N-methyl-D-aspartate
- NPY, neuropeptide Y
- Oxy, oxycodone
- PARV, parvalbumin
- PB, phosphate buffer
- PFA, paraformaldehyde
- PM, plasma membrane
- Pyramidal cells
- ROI, region of interest
- SLM, stratum lacunosum-moleculare
- SLu, stratum lucidum
- SO, stratum oriens
- SOM, somatostatin
- SR, stratum radiatum
- Sal, saline
- TS, tris-buffered saline
- ir, immunoreactivity
Collapse
Affiliation(s)
- Alexandra Dolgetta
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Megan Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Kate Fruitman
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Luke Siegel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
| | - Yan Zhou
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Bruce S. McEwen
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Mary Jeanne Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Teresa A. Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY, 10065, USA
- Harold and Margaret Milliken Hatch Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
48
|
Mental and substance use disorders in Peru: a systematic analysis of the Global Burden of Disease study. J Public Health (Oxf) 2022. [DOI: 10.1007/s10389-020-01341-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
49
|
Peart DR, Andrade AK, Logan CN, Knackstedt LA, Murray JE. Regulation of Cocaine-related Behaviors by Estrogen and Progesterone. Neurosci Biobehav Rev 2022; 135:104584. [DOI: 10.1016/j.neubiorev.2022.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/30/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
|
50
|
Kaplan G, Xu H, Abreu K, Feng J. DNA Epigenetics in Addiction Susceptibility. Front Genet 2022; 13:806685. [PMID: 35145550 PMCID: PMC8821887 DOI: 10.3389/fgene.2022.806685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/06/2022] [Indexed: 12/22/2022] Open
Abstract
Addiction is a chronically relapsing neuropsychiatric disease that occurs in some, but not all, individuals who use substances of abuse. Relatively little is known about the mechanisms which contribute to individual differences in susceptibility to addiction. Neural gene expression regulation underlies the pathogenesis of addiction, which is mediated by epigenetic mechanisms, such as DNA modifications. A growing body of work has demonstrated distinct DNA epigenetic signatures in brain reward regions that may be associated with addiction susceptibility. Furthermore, factors that influence addiction susceptibility are also known to have a DNA epigenetic basis. In the present review, we discuss the notion that addiction susceptibility has an underlying DNA epigenetic basis. We focus on major phenotypes of addiction susceptibility and review evidence of cell type-specific, time dependent, and sex biased effects of drug use. We highlight the role of DNA epigenetics in these diverse processes and propose its contribution to addiction susceptibility differences. Given the prevalence and lack of effective treatments for addiction, elucidating the DNA epigenetic mechanism of addiction vulnerability may represent an expeditious approach to relieving the addiction disease burden.
Collapse
|