1
|
Nadal-Gratacós N, Mata S, Puigseslloses P, De Macedo M, Lardeux V, Pain S, Wang FH, Källsten L, Pubill D, Berzosa X, Kehr J, Solinas M, Camarasa J, Escubedo E, López-Arnau R. Unveiling the potential abuse liability of α-D2PV: A novel α-carbon phenyl-substituted synthetic cathinone. Neuropharmacology 2025; 272:110425. [PMID: 40118207 DOI: 10.1016/j.neuropharm.2025.110425] [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: 01/28/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Synthetic cathinones are emerging psychoactive substances designed to mimic the effects of classical psychostimulants. Among them, α-D2PV, a novel pyrrolidine-containing cathinone characterized by a phenyl group on the α-carbon atom, has gained significant attention. This study investigates the in vitro and in silico mechanism of action as well as the abuse liability of α-D2PV using rodent models. Dopamine (DA), noradrenaline (NE), and serotonin (5-HT) uptake inhibition assays were conducted in HEK293 cells expressing the corresponding human monoamine transporter, complemented by molecular docking studies at the DA transporter (DAT). Behavioral studies in male Swiss CD-1 mice assessed locomotor activity and conditioned place preference, while microdialysis and self-administration experiments were performed in male Sprague-Dawley rats. The findings reveal that α-D2PV is a potent DA and NE uptake inhibitor, with minimal activity at the 5-HT transporter (SERT). Docking studies showed that the benzene rings of α-PVP and α-D2PV align precisely in their most stable conformations at DAT. In vivo, α-D2PV elicited dose-dependent hyperlocomotion, thigmotaxis, and rewarding effects in mice, alongside increased extracellular DA levels in the nucleus accumbens of awake rats. Self-administration experiments confirmed α-D2PV's high reinforcing efficacy, indicating a significant risk of abuse in humans. Finally, these results underscore the necessity for continued surveillance of α-D2PV within the illicit drug market. Furthermore, novel synthetic cathinones incorporating a phenyl ring at the α-carbon side chain warrant proactive monitoring due to their potential to retain dopaminergic activity and evade initial legal controls.
Collapse
Affiliation(s)
- Núria Nadal-Gratacós
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Sandra Mata
- Chemical Reactions for Innovative Solutions (CRISOL), IQS School of Engineering, Universitat Ramon Llull, 08017, Barcelona, Spain
| | - Pol Puigseslloses
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Morgane De Macedo
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| | - Virginie Lardeux
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| | - Stephanie Pain
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| | - Fu-Hua Wang
- Pronexus Analytical AB, Grindstuvägen 44, 167 33, Bromma, Sweden
| | | | - David Pubill
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Xavier Berzosa
- Chemical Reactions for Innovative Solutions (CRISOL), IQS School of Engineering, Universitat Ramon Llull, 08017, Barcelona, Spain
| | - Jan Kehr
- Pronexus Analytical AB, Grindstuvägen 44, 167 33, Bromma, Sweden
| | - Marcello Solinas
- Laboratoire de Neurosciences Expérimentales et Cliniques, Université de Poitiers, INSERM, U-1084, Poitiers, France
| | - Jordi Camarasa
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Elena Escubedo
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Raul López-Arnau
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Pharmacology Section and Institute of Biomedicine (IBUB), Faculty of Pharmacy, Universitat de Barcelona, 08028, Barcelona, Spain.
| |
Collapse
|
2
|
Kim MR, Haakenson CM, Brown JA, Ball GF, Prior NH. A social conditioned place preference test and pair maintenance in the monogamous zebra finch. Physiol Behav 2025:114925. [PMID: 40252942 DOI: 10.1016/j.physbeh.2025.114925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Social relationships are essential for a wide range of species, including humans. The mechanisms of pair formation and maintenance appear to be distinct, although relatively little is known about pair maintenance compared to bond formation. A social conditioned place preference (SCPP) test, a paradigm not often used, could be used to reveal differences in the mechanisms of bond formation and maintenance. Here we used the SCPP test across two experiments to investigate how the behavioral and neuroendocrine mechanisms of social affiliation change over the course of pair bonding in the monogamous zebra finch. In our procedure, we had three 30-minute phases: preconditioning occurring on Day 1, followed by conditioning, and postconditioning both on Day 2. In our first experiment, we simply asked whether courting and paired dyads would form SCPPs in this assay. We found evidence that courting, but not paired birds, formed SCPPs. This was particularly evident for courting females. Courting and paired dyads also engaged in different behaviors during the conditioning phase. Despite these effects of social context on behavior, there was no clear relationship between these behavioral differences and the formation of SCPPs for courting males or females. We next tested whether pharmacological treatment with D1 and D2 receptor agonists would alter the effects of social contexts on the formation of SCPPs. Specifically, we hypothesized that treatment with a D1-agonist (SKF 38393) would block the formation of SCPPs in courting dyads and that activation of D2-like receptors (quinpirole) would induce SCPPs in already established pairs. In contrast with Experiment 1, we did not see any groups forming SCPPs, but paired males, regardless of pharmacological treatment, appeared to form aversions to the conditioning side. Furthermore, there was significantly less courtship and affiliative behavior in the second experiment. Combined, this leads us to suspect that coupling the SCPP with a s.c. injection made the entire procedure too aversive to reveal any SCPP. Despite this, there were subtle effects of pharmacological manipulation on social behavior during the conditioning phase. All together, the results from our two experiments 1) suggest that there are differences in the mechanisms by which courting and paired dyads relate to their partner in novel environments, 2) highlight the differences in social interactions between courting and paired birds, and 3) leave open the possibility that dopamine signaling differentially shapes social behavior and the formation of SCPP in courting and paired birds.
Collapse
Affiliation(s)
- Martin R Kim
- Department of Psychology, Cornell University, Ithaca, NY
| | - Chelsea M Haakenson
- Neuroscience and Cognitive Science Program, Department of Psychology, University of Maryland, College Park, MD
| | - Jane A Brown
- Neuroscience and Cognitive Science Program, Department of Psychology, University of Maryland, College Park, MD
| | - Gregory F Ball
- Neuroscience and Cognitive Science Program, Department of Psychology, University of Maryland, College Park, MD
| | - Nora H Prior
- Department of Psychology, Cornell University, Ithaca, NY.
| |
Collapse
|
3
|
Gu X, Chen W, Li Z, Wang X, Su Q, Zhou F. Drp1 mitochondrial fission in astrocyte modulates behavior and neuroinflammation during morphine addiction. J Neuroinflammation 2025; 22:108. [PMID: 40247294 PMCID: PMC12007278 DOI: 10.1186/s12974-025-03438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/05/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Mitochondrial dynamics in neurons accompanied by neuroinflammation has been proved as pivotal events during repeated morphine exposure, however, the relationship between mitochondrial dynamics and neuroinflammation still remains unknown. METHODS This study was designed to investigate the potential role of astrocyte Drp1 in neuroinflammation during morphine addiction. Nucleus accumbens (NAc) tissues were collected for immunofluorescence, transmission electron microscopy (TEM) and quantitative real-time polymerase chain reaction (qRT-PCR) to detect the expression of pro-inflammatory cytokines and mitochondrial fission proteins. Morphine-induced conditioned place preference (CPP) and open field test (OFT) were used to determine the effects of Mdivi-1, a selective inhibitor of mitochondrial fission protein Drp1 in the rewarding properties of morphine. Astrocyte-specific knockdown experiments by an adeno-associated virus (AAV) vector containing shRNADrp1-EGFP infusion were performed to determine the effects of astrocyte Drp1 in NAc of mice with morphine treatment. RESULTS In this study, we found that repeated morphine exposure induced mitochondrial fragmentation in neurons, astrocytes, and microglia in NAc, correlating with increased inflammatory markers and addictive behaviors. The application of Mdivi-1 effectively mitigated mitochondrial fragmentation and astrocyte-mediated neuroinflammation within the NAc, thereby alleviating morphine-induced addictive behaviors. Crucially, the astrocyte-specific knockdown of Drp1 in NAc significantly curtailed drug-seeking behavior and substantially reduced neuroinflammation. CONCLUSIONS Collectively, our findings suggest that alterations in mitochondrial dynamics, particularly within astrocytes, play an important role in regulating neuroinflammation associated with morphine addiction. This research offers novel insights into potential therapeutic strategies for addressing substance use disorder (SUD) by regulating mitochondrial dynamics within astrocyte.
Collapse
Affiliation(s)
- Xiaotong Gu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China
| | - Wenjing Chen
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China
| | - Zixin Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China
| | - Xinran Wang
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China
| | - Qianying Su
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China
| | - Feifan Zhou
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya, 572025, China.
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Haikou, 570100, China.
| |
Collapse
|
4
|
Nguyen CH, Salazar MG, Rincón-Cortés M. Female rats exposed to early life scarcity-adversity are resilient to later life changes in maternal behavior. Horm Behav 2025; 170:105720. [PMID: 40090292 DOI: 10.1016/j.yhbeh.2025.105720] [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: 07/02/2024] [Revised: 01/10/2025] [Accepted: 03/08/2025] [Indexed: 03/18/2025]
Abstract
In both humans and rodents, maternal care is disturbed by exposure to environmental adversity, including low resource conditions (i.e., poverty, scarcity). Maternal adversity is associated with compromised quality of mother-infant attachment and increased adverse caregiving patterns such as abuse, maltreatment and/or neglect, which disrupt behavioral development in the female offspring. Importantly, maternal behavior is thought to be an intergenerational behavior, meaning that the quality of maternal care a female experiences during early life is thought to influence the quality of care she will display towards her own offspring when she becomes a mother. Here, we tested this idea by employing a rodent model of postpartum environmental adversity based on creating an impoverished nesting environment during postpartum days (PD) 2-9, which disrupts mother-infant interactions and is thought to upregulate hypothalamic-pituitary-adrenal (HPA)-axis function in the pups. We examined the impact of this form of early life adversity on pup stress hormone (i.e., corticosterone- CORT) levels by collecting trunk blood and later life maternal behavior by conducting maternal behavior observations and maternal motivation tests (e.g., T-Maze, pup retrieval, pup-associated conditioned place preference) in the first filial (F1) generation. We report no impact of early life scarcity-adversity/adverse caregiving on pup CORT levels or later life naturalistic or motivated maternal behaviors. In sum, we show that female rat pups who experienced adverse caregiving during early life showed resilience towards developing negative caregiving patterns, as they did not perpetuate the same aberrant maternal behavior that they received from their mothers.
Collapse
Affiliation(s)
- Christine H Nguyen
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Melissa G Salazar
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Millie Rincón-Cortés
- Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
5
|
Ruyle BC, Masud S, Kesaraju R, Tahirkheli M, Modh J, Roth CG, Angulo-Lopera S, Lintz T, Higginbotham JA, Massaly N, Morón JA. Peripheral opioid receptor antagonism alleviates fentanyl-induced cardiorespiratory depression and is devoid of aversive behavior. eLife 2025; 13:RP104469. [PMID: 40167443 PMCID: PMC11961120 DOI: 10.7554/elife.104469] [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] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Millions of Americans suffering from Opioid Use Disorders face a high risk of fatal overdose due to opioid-induced respiratory depression (OIRD). Fentanyl, a powerful synthetic opioid, is a major contributor to the rising rates of overdose deaths. Reversing fentanyl overdoses has proved challenging due to its high potency and the rapid onset of OIRD. We assessed the contributions of central and peripheral mu opioid receptors (MORs) in mediating fentanyl-induced physiological responses. The peripherally restricted MOR antagonist naloxone methiodide (NLXM) both prevented and reversed OIRD to a degree comparable to that of naloxone (NLX), indicating substantial involvement of peripheral MORs to OIRD. Interestingly, NLXM-mediated OIRD reversal did not produce aversive behaviors observed after NLX. We show that neurons in the nucleus of the solitary tract (nTS), the first central synapse of peripheral afferents, exhibit a biphasic activity profile following fentanyl exposure. NLXM pretreatment attenuates this activity, suggesting that these responses are mediated by peripheral MORs. Together, these findings establish a critical role for peripheral MORs, including ascending inputs to the nTS, as sites of dysfunction during OIRD. Furthermore, selective peripheral MOR antagonism could be a promising therapeutic strategy for managing OIRD by sparing CNS-driven acute opioid-associated withdrawal and aversion observed after NLX.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Sarah Masud
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Rohith Kesaraju
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Mubariz Tahirkheli
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Juhi Modh
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Caroline G Roth
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Sofia Angulo-Lopera
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Tania Lintz
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Jessica A Higginbotham
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Nicolas Massaly
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
| | - Jose A Morón
- Department of Anesthesiology, Washington University in St LouisSt LouisUnited States
- Pain Center, Washington University in St LouisSt LouisUnited States
- School of Medicine, Washington University in St. LouisSt. LouisUnited States
- Department of Neuroscience, Washington University in St. LouisSt. LouisUnited States
- Department of Psychiatry, Washington University in St. LouisSt. LouisUnited States
| |
Collapse
|
6
|
Plewnia C, Masini D, Fisone G. Rewarding properties of L-Dopa in experimental parkinsonism are mediated by sensitized dopamine D1 receptors in the dorsal striatum. Mol Psychiatry 2025; 30:976-985. [PMID: 39227434 PMCID: PMC11835726 DOI: 10.1038/s41380-024-02721-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/05/2024]
Abstract
Treatment of Parkinson's disease (PD) is based on the use of dopaminergic drugs, such as L-Dopa and dopamine receptor agonists. These substances counteract motor symptoms, but their administration is accompanied by motor and non-motor complications. Among these latter conditions a neurobehavioral disorder similar to drug abuse, known as dopamine dysregulation syndrome (DDS), is attracting increasing interest because of its profound negative impact on the patients' quality of life. Here we replicate DDS in a PD mouse model based on a bilateral injection of 6-hydroxydopamine (6-OHDA) into the dorsal striatum. Administration of L-Dopa induced locomotor sensitization and conditioned place preference in 6-OHDA lesion, but not in control mice, indicative of the acquisition of addictive-like properties following nigrostriatal dopamine depletion. These behavioral effects were accompanied by abnormal dopamine D1 receptor (D1R) signaling in the medium spiny neurons of the dorsal striatum, leading to hyperactivation of multiple signaling cascades and increased expression of ΔFosB, a stable transcription factor involved in addictive behavior. Systemic administration of the D1R antagonist, SCH23390, abolished these effects and the development of place preference, thereby counteracting the psychostimulant-like effect of L-Dopa. The rewarding properties of L-Dopa were also prevented by chemogenetic inactivation of D1R-expressing neurons in the dorsal striatum. Our results indicate the association between abnormal D1R-mediated transmission and DDS in PD and identify potential approaches for the treatment of this disorder.
Collapse
Affiliation(s)
- Carina Plewnia
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
| | - Débora Masini
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, 10691, Stockholm, Sweden
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, 17177, Stockholm, Sweden.
| |
Collapse
|
7
|
Day AW, Perez-Lozada J, DiLeo A, Blandino K, Maguire J, Kumamoto CA. Candida albicans Colonization Modulates Murine Ethanol Consumption and Behavioral Responses Through Elevation of Serum Prostaglandin E 2 and Impact on the Striatal Dopamine System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640044. [PMID: 40060518 PMCID: PMC11888247 DOI: 10.1101/2025.02.25.640044] [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: 03/14/2025]
Abstract
Candida albicans is a commensal yeast that is a common component of the gastrointestinal (GI) microbiome of humans. C. albicans has been shown to bloom in the GI tract of individuals with alcohol use disorder (AUD) and can promote and increase the severity of alcoholic liver disease (ALD). However, the effects of C. albicans blooms on the host in the context of AUD or AUD-related phenotypes, such as ethanol preference, have been unstudied. In this work, we report a reduction in ethanol consumption and preference in mice colonized with C. albicans. C. albicans-colonized mice exhibited elevated levels of serum PGE2 and reduced ethanol preference was reversed by injection with antagonists of PGE2 receptors. Further, injection of mice with a PGE2 derivative decreased their ethanol preference. These results show that PGE2 acting on its receptors EP1 and EP2 drives reduced ethanol preference in C. albicans-colonized mice. We also showed altered transcription of dopamine receptors in the dorsal striatum of C. albicans-colonized mice and more rapid acquisition of ethanol conditioned taste aversion, suggesting alterations to reinforcement or aversion learning. Finally, C. albicans-colonized mice were more susceptible to ethanol-induced motor coordination impairment showing significant alterations to the behavioral effects of ethanol. This study identifies a member of the fungal microbiome that alters ethanol preference and demonstrates a role for PGE2 signaling in these phenotypes.
Collapse
Affiliation(s)
- Andrew W. Day
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jeyra Perez-Lozada
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
- current address: San Juan Bautista School of Medicine, Caguas, Puerto Rico, 00727, USA
| | - Alyssa DiLeo
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Katrina Blandino
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jamie Maguire
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| |
Collapse
|
8
|
He Y, Piao ZS, Jia Y, Wu HQ, Wang XQ, Yu WF, Wu FX. Dezocine modulates the reinstatement of conditioned place preference in morphine-dependent rats via the dopamine reward circuitry. Front Neurosci 2025; 19:1507747. [PMID: 40040846 PMCID: PMC11876162 DOI: 10.3389/fnins.2025.1507747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Opioid addiction is a significant public health issue, with existing treatments such as buprenorphine and methadone exhibiting limitations, including side effects and insufficient prevention of relapse. Novel therapeutic strategies are needed to address these challenges. This study investigates the potential of dezocine in reducing addiction-related behaviors and preventing relapse. Methods A morphine-induced conditioned place preference (CPP) model was established in rats to evaluate the effect of dezocine on addiction-related behaviors. Behavioral assessments were conducted to measure withdrawal symptoms and CPP reinstatement. To explore the underlying mechanism, Western blot (WB) and immunofluorescence (IF) were used to quantify the expression of phosphorylated DARPP32 (p-DARPP32) and DOPA decarboxylase (DDC) in reward-related brain regions, including the nucleus accumbens (NAc), ventral tegmental area (VTA), hippocampus (HP), and prefrontal cortex (PFC). Results Dezocine significantly reduced withdrawal symptoms and prevented CPP reinstatement, indicating its potential to alleviate addiction behaviors. Western blotting and immunofluorescence analysis revealed that dezocine increased p-DARPP32 expression in the NAc, VTA, HP, and PFC, without altering DDC levels. Discussion These findings suggest that dezocine may exert its therapeutic effects by inhibiting kappa opioid receptor activation and enhancing dopamine signaling in reward-related brain circuitry. The increase in p-DARPP32 expression in key brain regions supports this mechanism, providing insights into the potential clinical application of dezocine for managing opioid addiction. Dezocine represents a promising candidate for opioid addiction treatment, with the ability to control withdrawal symptoms and prevent relapse.
Collapse
Affiliation(s)
- Yan He
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
- Department of Anesthesiology, Fuzhou First Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Zhi-Sheng Piao
- Department of Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Yi Jia
- Department of Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Hui-Qi Wu
- Department of Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
| | - Xiao-Qiang Wang
- Department of Anesthesiology, Ren Ji Hospital of Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
- Department of Anesthesiology, Ren Ji Hospital of Shanghai Jiao Tong University, Shanghai, China
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fei-Xiang Wu
- Department of Critical Care Medicine, Eastern Hepatobiliary Surgery Hospital Affiliated to Naval Medical University, Shanghai, China
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
9
|
Amawi H, Hammad AM, Ibrahim AA, Alsbih N, Hall FS, Alasmari F, Al-Trad B. Impact of Exercise on Tramadol-Conditioned Place Preference. Brain Sci 2025; 15:89. [PMID: 39851456 PMCID: PMC11763564 DOI: 10.3390/brainsci15010089] [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: 12/07/2024] [Revised: 01/12/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Tramadol (TRA) is an opioid that is used to manage moderate to severe pain. Long-term use of TRA can lead to the development of opioid use disorder. OBJECTIVES This study investigates the role of forced exercise in reducing TRA-seeking behavior. METHODS Adult male rats (240-260 g) were divided into five groups; the control group received vehicle injections, the TRA group received TRA (75 mg/kg, i.p) every other day for 8 days, and three TRA-exercise groups were forced to run on a treadmill (60 min/day, 5 days/week) for 2, 4, or 6 weeks prior to conditioning with TRA. A tramadol-conditioned place preference (CPP) procedure assessed TRA reinforcement, after which all rats were euthanized, tissue extracted, and mRNA expression for brain-derived neurotrophic factor (Bdnf) and interleukin 1 beta (Il-1β) determined in hippocampus (Hipp), prefrontal cortex (PFC), and nucleus accumbens (NAc). RESULTS TRA-seeking behavior was seen in the TRA group and the 6 weeks forced exercise group. By contrast, forced exercise for 2 or 4 weeks attenuated TRA-seeking behavior. This attenuation was associated with a significant increase in Bdnf mRNA expression in the Hipp and NAc, but not the PFC. Additionally, the TRA-induced elevations in Il-1β mRNA expression were reversed by all durations of exercise in Hipp. However, only 2 and 4 weeks, but not 6 weeks, of exercise reduced elevations in PFC and NAc Il-1β expression. CONCLUSION Forced exercise for 2 and 4 weeks attenuates TRA-seeking behavior partially through the regulation of Bdnf and Il-1β mRNA expression.
Collapse
Affiliation(s)
- Haneen Amawi
- Department of Clinical Pharmacy and Pharmacy Practice, College of Pharmacy, Yarmouk University, Irbid 21163, Jordan
| | - Alaa M. Hammad
- Department of Pharmacy, College of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Aseel Abdullah Ibrahim
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan (B.A.-T.)
| | - Nosyba Alsbih
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan (B.A.-T.)
| | - Frank Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA;
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Bahaa Al-Trad
- Department of Biological Sciences, Faculty of Science, Yarmouk University, Irbid 21163, Jordan (B.A.-T.)
| |
Collapse
|
10
|
Ruyle BC, Masud S, Kesaraju R, Tahirkheli M, Modh J, Roth CG, Angulo-Lopera S, Lintz T, Higginbotham JA, Massaly N, Moron JA. Peripheral opioid receptor antagonism alleviates fentanyl-induced cardiorespiratory depression and is devoid of aversive behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.16.613257. [PMID: 39345613 PMCID: PMC11429738 DOI: 10.1101/2024.09.16.613257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Millions of Americans suffering from Opioid Use Disorders (OUD) face a high risk of fatal overdose due to opioid-induced respiratory depression (OIRD). Fentanyl, a powerful synthetic opioid, is a major contributor to the rising rates of overdose deaths. Reversing fentanyl overdoses has proved challenging due to its high potency and the rapid onset of OIRD. We assessed the contributions of central and peripheral mu opioid receptors (MORs) in mediating fentanyl-induced physiological responses. The peripherally restricted MOR antagonist naloxone methiodide (NLXM) both prevented and reversed OIRD to a degree comparable to that of naloxone (NLX), indicating substantial involvement of peripheral MORs to OIRD. Interestingly, NLXM-mediated OIRD reversal did not produce aversive behaviors observed after NLX. We show that neurons in the nucleus of the solitary tract (nTS), the first central synapse of peripheral afferents, exhibit a biphasic activity profile following fentanyl exposure. NLXM pretreatment attenuates this activity, suggesting that these responses are mediated by peripheral MORs. Together, these findings establish a critical role for peripheral MORs, including ascending inputs to the nTS, as sites of dysfunction during OIRD. Furthermore, selective peripheral MOR antagonism could be a promising therapeutic strategy for managing OIRD by sparing CNS-driven acute opioid-associated withdrawal and aversion observed after NLX. Significance Statement In this study, we compare the central versus peripheral components underlying fentanyl-induced cardiorespiratory depression to prevent overdose deaths. Our data indicate that these effects are, at least partially, due to the activation of mu opioid receptors present in peripheral sites. These findings provide insight into peripheral contributions to fentanyl-induced overdoses and could potentially lead to the development of treatments selectively targeting the peripheral system, sparing individuals from the CNS-driven acute opioid withdrawal generally observed with the use of naloxone.
Collapse
|
11
|
Moran KM, Jarrell L, Khashchuluun M, Moran KR, Rodriguez J, Tran A, Delville Y. Blunted food conditioned place preference-like behavior in adolescent-stressed male hamsters. Behav Brain Res 2025; 476:115234. [PMID: 39233144 DOI: 10.1016/j.bbr.2024.115234] [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: 05/17/2024] [Revised: 08/01/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
Social stress during adolescence results in long lasting weight gain, obesity, and enhanced food hoarding behavior in hamsters. We wanted to determine whether stress also enhanced conditioned place preference-like behavior (CPP-like) for food reward, as would be expected from studies with substances like cocaine. Our experimental animals were exposed daily to aggressive adults for two weeks in early puberty, while also trained to explore a V-shaped maze containing a food reward at one end. They were tested for CPP-like behavior on the last day of social stress. Our results showed that while stress enhanced weight gain, food intake, food efficiency, and body fat, it caused a reduction of Place Preference as compared to controls. In fact, the correlated relationship between Place Preference and body fat was inverted by stress exposure: while it was positively correlated in controls, it was mildly negatively correlated in stressed hamsters. These unexpected data illustrate the extent of adaptive behavior in foraging animals once a resource has become untrustworthy.
Collapse
Affiliation(s)
- Kevin M Moran
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Leah Jarrell
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA
| | | | - Kurt R Moran
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA
| | - Julia Rodriguez
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA
| | - Anna Tran
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yvon Delville
- Psychology Department, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
12
|
Kundu D, Acharya S, Wang S, Cao Y, Kim HJ, Cheong JH, Kim KM. Roles of metabotropic signaling of nicotine receptors in the development and maintenance of nicotine reward through regulation of dopamine D 3 receptor expression. J Neurochem 2025; 169:e16271. [PMID: 39696743 DOI: 10.1111/jnc.16271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/03/2024] [Accepted: 11/11/2024] [Indexed: 12/20/2024]
Abstract
The α4β2 nicotinic acetylcholine receptor (nAChR), an ionophore, has been suggested to signal through metabotropic pathways and interact with other receptor families, such as dopamine receptors. In this study, the interaction between α4β2 nAChR and dopamine receptors was investigated through in vivo and in vitro studies. Nicotine exposure in adolescent rats is known to induce a sustained increase in nicotine's rewarding effects which was assessed by conditioned place preference (CPP) assay. The expression levels of α4β2 nAChR and dopamine D2/D3 receptors (D2R, D3R) increased after nicotine treatment. To determine which of these two dopamine receptors was increased by nicotine treatment, a newly developed ligand with high selectivity for D3R was used in the radioligand binding assay. Although the expression of both α4β2 nAChR and D3R was enhanced by nicotine exposure during adolescence, only the elevated level of D3R persisted into adulthood. In experiments conducted on mice, D3R knockout mice showed significantly lower CPP scores in adulthood compared to wild-type mice. Cellular studies showed that an increase in D3R expression was attributed to enhanced D3R promoter activity, regulated by a signaling cascade composed of Src, Syk, PKC, and NF-κB. These results demonstrate that the metabotropic signaling pathway is involved in the interaction between α4β2 nAChR and D3R, and also suggest how nicotine reward initiated in adolescence could relapse after a long abstinence period. Given the significance of adolescent nicotine exposure on nicotine addiction, this study is thought to offer a novel mechanistic perspective for understanding nicotine reward and relapse.
Collapse
Affiliation(s)
- Dooti Kundu
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Srijan Acharya
- St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Shujie Wang
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Yongkai Cao
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Hee Jin Kim
- College of Pharmacy, Sahmyook University, Seoul, Republic of Korea
| | - Jae Hoon Cheong
- College of Pharmacy, Jeonbuk National University, Jeonju, Republic of Korea
| | - Kyeong-Man Kim
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
13
|
Chen Z, Tang S, Xiao X, Hong Y, Fu B, Li X, Shao Y, Chen L, Yuan D, Long Y, Wang H, Hong H. Adiponectin receptor 1-mediated basolateral amygdala-prelimbic cortex circuit regulates methamphetamine-associated memory. Cell Rep 2024; 43:115074. [PMID: 39661515 DOI: 10.1016/j.celrep.2024.115074] [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: 06/19/2024] [Revised: 10/14/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
The association between drug-induced rewards and environmental cues represents a promising strategy to address addiction. However, the neural networks and molecular mechanisms orchestrating methamphetamine (MA)-associated memories remain incompletely characterized. In this study, we demonstrated that AdipoRon (AR), a specific adiponectin receptor (AdipoR) agonist, inhibits the formation of MA-induced conditioned place preference (CPP) in MA-conditioned mice, accompanied by suppression of basolateral amygdala (BLA) CaMKIIα neuron activity. Furthermore, we identified an association between the excitatory circuit from the BLA to the prelimbic cortex (PrL) and the integration of MA-induced rewards with environmental cues. We also determined that the phosphorylated AMPK (p-AMPK)/Cav1.3 signaling pathway mediates the modulatory effects of AdipoR1 in PrL-projecting BLA CaMKIIα neurons on the formation of MA reward memories, a process influenced by physical exercise. These findings highlight the critical function of AdipoR1 in the BLACaMKIIα→PrLCaMKIIα circuit in regulating MA-related memory formation, suggesting a potential target for managing MA use disorders.
Collapse
Affiliation(s)
- Zhigang Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Susu Tang
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyi Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yizhou Hong
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Boli Fu
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuyi Li
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yuwei Shao
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Chen
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Danhua Yuan
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Long
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine/Nanhu Brain-computer Interface Institute, Hangzhou 310013, China.
| | - Hao Hong
- College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
14
|
Isaac J, Murugan M. Interconnected neural circuits mediating social reward. Trends Neurosci 2024; 47:1041-1054. [PMID: 39532581 PMCID: PMC11633286 DOI: 10.1016/j.tins.2024.10.004] [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: 05/17/2024] [Revised: 08/26/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Across species, social behaviors are shaped and maintained through positive reinforcement of affiliative social interactions. As with nonsocial rewards, the reinforcing properties of social interactions have been shown to involve interplay between various brain regions and the mesolimbic reward system. In this review, we summarize findings from rodent research on the neural circuits that encode and mediate different components of social reward-seeking behavior. We explore methods to parse and study social reward-related behaviors using available behavioral paradigms. We also compare the neural mechanisms that support social versus nonsocial reward-seeking. Finally, we discuss how internal state and neuromodulatory systems affect reward-seeking behavior and the neural circuits that underlie social reward.
Collapse
Affiliation(s)
- Jennifer Isaac
- Neuroscience Graduate Program, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Malavika Murugan
- Neuroscience Graduate Program, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
15
|
Martínez-Rivera A, Fetcho RN, Birmingham L, Xu J, Yang R, Foord C, Scala-Chávez D, Mekawy N, Pleil K, Pickel VM, Liston C, Castorena CM, Levitz J, Pan YX, Briand LA, Rajadhyaksha AM, Lee FS. Elevating levels of the endocannabinoid 2-arachidonoylglycerol blunts opioid reward but not analgesia. SCIENCE ADVANCES 2024; 10:eadq4779. [PMID: 39612328 PMCID: PMC11606496 DOI: 10.1126/sciadv.adq4779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
Converging findings have established that the endocannabinoid (eCB) system serves as a possible target for the development of new treatments as a complement to opioid-based treatments. Here, we show in male and female mice that enhancing levels of the eCB, 2-arachidonoylglycerol (2-AG), through pharmacological inhibition of its catabolic enzyme, monoacylglycerol lipase (MAGL), either systemically or in the ventral tegmental area (VTA) with JZL184, leads to a substantial attenuation of the rewarding effects of opioids in mice using conditioned place preference and self-administration paradigms, without altering their analgesic properties. These effects are driven by cannabinoid receptor 1 (CB1R) within the VTA, as VTA CB1R conditional knockout counteracts JZL184's effects. Using fiber photometry with fluorescent sensors for calcium and dopamine (DA), we find that enhancing 2-AG levels diminishes opioid reward-related nucleus accumbens (NAc) activity and DA neurotransmission. Together, these findings reveal that 2-AG diminishes the rewarding properties of opioids and provides a potential adjunctive therapeutic strategy for opioid-related analgesic treatments.
Collapse
Affiliation(s)
- Arlene Martínez-Rivera
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Robert N. Fetcho
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lizzie Birmingham
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Jin Xu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Ruirong Yang
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Careen Foord
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Diego Scala-Chávez
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Narmin Mekawy
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kristen Pleil
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Virginia M. Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Conor Liston
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carlos M. Castorena
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lisa A. Briand
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122, USA
| | - Anjali M. Rajadhyaksha
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Center for Substance Abuse Research and Department of Neural Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Francis S. Lee
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
16
|
Wang Y, Guo S, Ventura T, Herzig V. Using a conditioned place preference assay in fruit flies to examine effects of insecticidal compounds on contextual memory. Toxicon 2024; 251:108128. [PMID: 39413976 DOI: 10.1016/j.toxicon.2024.108128] [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: 08/16/2024] [Revised: 09/30/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
Insecticides are vital for safeguarding agricultural crops against pests, albeit many lack selectivity towards pest species and are poorly bio-degradable. This leads to targeting of beneficial organisms like pollinators and widespread environmental contamination of soil and water. Exposure to insecticides such as neonicotinoids causes insect paralysis and mortality at higher doses, while sublethal doses can disrupt other functions that are crucial for survival such as learning and memory performance. Potent and selective arachnid venom peptides affecting a variety of molecular targets are being explored as bioinsecticide candidates. However, their effect on insect learning is poorly understood. We therefore established a sucrose-induced conditioned place preference (CPP) assay using Drosophila melanogaster fruit flies to provide a means of evaluating how various classes of insecticidal compounds interact with insect memory to assess their broader ecological consequences. Our results confirmed the adverse effect of a sublethal dose of the neonicotinoid insecticide imidacloprid (20 pg/fly) on fly CPP formation upon daily injection during the conditioning phase. However, imidacloprid did not affect CPP retrieval when applied after the conditioning phase. Sublethal doses of the two insecticidal spider venom peptides μ-DGTX-Dc1a (Dc1a; 70 pg/fly) and U1-AGTX-Ta1a (Ta1a; 125 pg/fly) had no effect on either CPP formation or retrieval, underlining their potential as novel and safe bioinsecticide candidates.
Collapse
Affiliation(s)
- Yachen Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Shaodong Guo
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Tomer Ventura
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia
| | - Volker Herzig
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD, 4556, Australia.
| |
Collapse
|
17
|
Barbano MF, Qi J, Chen E, Mohammad U, Espinoza O, Candido M, Wang H, Liu B, Hahn S, Vautier F, Morales M. VTA glutamatergic projections to the nucleus accumbens suppress psychostimulant-seeking behavior. Neuropsychopharmacology 2024; 49:1905-1915. [PMID: 38926603 PMCID: PMC11473768 DOI: 10.1038/s41386-024-01905-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024]
Abstract
Converging evidence indicates that both dopamine and glutamate neurotransmission within the nucleus accumbens (NAc) play a role in psychostimulant self-administration and relapse in rodent models. Increased NAc dopamine release from ventral tegmental area (VTA) inputs is critical to psychostimulant self-administration and NAc glutamate release from prelimbic prefrontal cortex (PFC) inputs synapsing on medium spiny neurons (MSNs) is critical to reinstatement of psychostimulant-seeking after extinction. The regulation of the activity of MSNs by VTA dopamine inputs has been extensively studied, and recent findings have demonstrated that VTA glutamate neurons target the NAc medial shell. Here, we determined whether the mesoaccumbal glutamatergic pathway plays a role in psychostimulant conditioned place preference and self-administration in mice. We used optogenetics to induce NAc release of glutamate from VTA inputs during the acquisition, expression, and reinstatement phases of cocaine- or methamphetamine-induced conditioned place preference (CPP), and during priming-induced reinstatement of cocaine-seeking behavior. We found that NAc medial shell release of glutamate resulting from the activation of VTA glutamatergic fibers did not affect the acquisition of cocaine-induced CPP, but it blocked the expression, stress- and priming-induced reinstatement of cocaine- and methamphetamine CPP, as well as it blocked the priming-induced reinstatement of cocaine-seeking behavior after extinction. These findings indicate that in contrast to the well-recognized mesoaccumbal dopamine system that is critical to psychostimulant reward and relapse, there is a parallel mesoaccumbal glutamatergic system that suppresses reward and psychostimulant-seeking behavior.
Collapse
Affiliation(s)
- M Flavia Barbano
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Jia Qi
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Emma Chen
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Uzma Mohammad
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Orlando Espinoza
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marcos Candido
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Huiling Wang
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Bing Liu
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Suyun Hahn
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - François Vautier
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Marisela Morales
- Integrative Neuroscience Research Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
18
|
Yin J, Zhang T, Li D, Xu F, Li H, Pan X, Liu F, Zhao Y, Weng X. Behavioral video coding analysis of chronic morphine administration in rats. Biomed Rep 2024; 21:168. [PMID: 39345955 PMCID: PMC11428083 DOI: 10.3892/br.2024.1856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/20/2024] [Indexed: 10/01/2024] Open
Abstract
The present study assessed the behavior of morphine-addicted rats using behavioral video coding technology, to evaluate effective methods for identifying morphine addiction. Rats were divided into a control group (n=15) and a morphine addiction group (n=15). The morphine addiction model was established with a 14-day increasing dose scheme, confirmed using a conditional place preference (CPP) experiment. After successful modeling, the rats' behavior was recorded for 12 h, then coded and analyzed using Observer XT behavior analysis software. Compared with the control group, morphine-addicted rats showed increased heat pain tolerance time (P=0.039) and spent more time in the white box during the CPP experiment (P<0.001). Video coding analysis revealed significant behavioral changes in morphine-addicted rats compared to controls. In addition to being lighter, morphine-addicted rats showed decreased water intake, reduced licking of forelimbs and hind limbs, and altered sleeping posture (sleeping curled up) during the day (all P<0.05). In conclusion, chronic morphine administration in rats leads to distinctive behavioral changes, including decreased licking frequency, reduced water intake and altered sleep posture. Video coding analysis, as a safe and non-invasive method, may provide a convenient and efficient approach for studying morphine addiction in rats.
Collapse
Affiliation(s)
- Jie Yin
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Tiecheng Zhang
- Department of Public Health, Chengdu Medical College, Sichuan 610500, P.R. China
| | - Dan Li
- Jingnan Medical Area of the General Hospital of the People's Liberation Army, Beijing 100850, P.R. China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Sichuan 610500, P.R. China
| | - Huan Li
- Department of Public Health, Chengdu Medical College, Sichuan 610500, P.R. China
| | - Xinyu Pan
- Department of Public Health, Chengdu Medical College, Sichuan 610500, P.R. China
| | - Fang Liu
- Department of Public Health, Chengdu Medical College, Sichuan 610500, P.R. China
| | - Yongqi Zhao
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| | - Xiechuan Weng
- Department of Neuroscience, Beijing Institute of Basic Medical Sciences, Beijing 100850, P.R. China
| |
Collapse
|
19
|
Fujikawa R, Ramsaran AI, Guskjolen A, de la Parra J, Zou Y, Mocle AJ, Josselyn SA, Frankland PW. Neurogenesis-dependent remodeling of hippocampal circuits reduces PTSD-like behaviors in adult mice. Mol Psychiatry 2024; 29:3316-3329. [PMID: 38719894 DOI: 10.1038/s41380-024-02585-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 11/08/2024]
Abstract
Post-traumatic stress disorder (PTSD) is a hypermnesic condition that develops in a subset of individuals following exposure to severe trauma. PTSD symptoms are debilitating, and include increased anxiety, abnormal threat generalization, and impaired extinction. In developing treatment strategies for PTSD, preclinical studies in rodents have largely focused on interventions that target post-encoding memory processes such as reconsolidation and extinction. Instead, here we focus on forgetting, another post-encoding process that regulates memory expression. Using a double trauma murine model for PTSD, we asked whether promoting neurogenesis-mediated forgetting can weaken trauma memories and associated PTSD-relevant behavioral phenotypes. In the double trauma paradigm, consecutive aversive experiences lead to a constellation of behavioral phenotypes associated with PTSD including increases in anxiety-like behavior, abnormal threat generalization, and deficient extinction. We found that post-training interventions that elevate hippocampal neurogenesis weakened the original trauma memory and decreased these PTSD-relevant phenotypes. These effects were observed using multiple methods to manipulate hippocampal neurogenesis, including interventions restricted to neural progenitor cells that selectively promoted integration of adult-generated granule cells into hippocampal circuits. The same interventions also weakened cocaine place preference memories, suggesting that promoting hippocampal neurogenesis may represent a broadly useful approach in hypermnesic conditions such as PTSD and substance abuse disorders.
Collapse
Affiliation(s)
- Risako Fujikawa
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Adam I Ramsaran
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
| | - Axel Guskjolen
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Juan de la Parra
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Yi Zou
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Andrew J Mocle
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
| | - Sheena A Josselyn
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Paul W Frankland
- Program in Neurosciences & Mental Health, Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, M5S 3G3, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5G 1X8, Canada.
- Institute of Medical Sciences, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Child & Brain Development Program, Canadian Institute for Advanced Research, Toronto, ON, M5G 1M1, Canada.
| |
Collapse
|
20
|
Borruto AM, Calpe-López C, Spanagel R, Bernardi RE. Conditional deletion of the AMPA-GluA1 and NMDA-GluN1 receptor subunit genes in midbrain D1 neurons does not alter cocaine reward in mice. Neuropharmacology 2024; 258:110081. [PMID: 39002853 DOI: 10.1016/j.neuropharm.2024.110081] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Synaptic plasticity in the mesolimbic dopamine (DA) system contributes to the neural adaptations underlying addictive behaviors and relapse. However, the specific behavioral relevance of glutamatergic excitatory drive onto dopamine D1 receptor (D1R)-expressing neurons in mediating the reinforcing effect of cocaine remains unclear. Here, we investigated how midbrain AMPAR and NMDAR function modulate cocaine reward-related behavior using mutant mouse lines lacking the glutamate receptor genes Gria1 or Grin1 in D1R-expressing neurons (GluA1D1CreERT2 or GluN1D1CreERT2, respectively). We found that conditional genetic deletion of either GluA1 or GluN1 within this neuronal sub-population did not impact the ability of acute cocaine injection to increase intracranial self-stimulation (ICSS) ratio or reduced brain reward threshold compared to littermate controls. Additionally, our data demonstrate that deletion of GluA1 and GluN1 receptor subunits within D1R-expressing neurons did not affect cocaine reinforcement in an operant self-administration paradigm, as mutant mice showed comparable cocaine responses and intake to controls. Given the pivotal role of glutamate receptors in mediating relapse behavior, we further explored the impact of genetic deletion of AMPAR and NMDAR onto D1R-expressing neurons on cue-induced reinstatement following extinction. Surprisingly, deletion of AMPAR and NMDAR onto these neurons did not impair cue-induced reinstatement of cocaine-seeking behavior. These findings suggest that glutamatergic activity via NMDAR and AMPAR in D1R-expressing neurons may not exclusively mediate the reinforcing effects of cocaine and cue-induced reinstatement.
Collapse
MESH Headings
- Animals
- Cocaine/pharmacology
- Cocaine/administration & dosage
- Receptors, N-Methyl-D-Aspartate/genetics
- Receptors, N-Methyl-D-Aspartate/metabolism
- Reward
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Mice
- Self Administration
- Male
- Mesencephalon/metabolism
- Mesencephalon/drug effects
- Conditioning, Operant/drug effects
- Conditioning, Operant/physiology
- Neurons/metabolism
- Neurons/drug effects
- Mice, Knockout
- Dopamine Uptake Inhibitors/pharmacology
- Mice, Inbred C57BL
- Reinforcement, Psychology
- Nerve Tissue Proteins
Collapse
Affiliation(s)
- Anna Maria Borruto
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | - Claudia Calpe-López
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; German Center for Mental Health (DZPG), Partner Site Mannheim, Heidelberg, Ulm, Germany
| | - Rick E Bernardi
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
21
|
Ede T, Ibach S, Parsons TD. Place conditioning as evaluation of affective valence in piglets. Sci Rep 2024; 14:25216. [PMID: 39448650 PMCID: PMC11502741 DOI: 10.1038/s41598-024-75849-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
The emotional valence of animals is challenging to assess, despite being a key component of welfare. In this study, we attempted to assess emotional valence through memory in 1- and 3-week-old piglets. It was hypothesized that piglets would spend less time in a pen where they experienced a negative event (castration) and more time in a pen where they experienced a positive event (enrichment). A testing apparatus was designed with three equally sized pens: two outer sections serving as treatment pens containing unique visual and tactile cues and a center section remaining neutral. Piglets received either negative or positive condition in one outer treatment pen and a sham treatment in the opposite. Various methods were tested (age of piglets, number and length of conditioning sessions, passive vs. active conditioning). Contrary to expectations, piglets did not decrease their time in the pen associated with the negative condition or increase their time in the pen associated with the positive condition. However, when exposed to the positive condition, results indicate older piglets developed an aversion towards the sham treatment. This study provides methodological groundwork for the application of place conditioning in piglets and highlights the nuances important for the use of cognitive tests to assess animal welfare.
Collapse
Affiliation(s)
- Thomas Ede
- Swine Teaching and Research Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA, USA.
| | - Sarah Ibach
- Swine Teaching and Research Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA, USA
| | - Thomas D Parsons
- Swine Teaching and Research Center, University of Pennsylvania School of Veterinary Medicine, Kennett Square, PA, USA
| |
Collapse
|
22
|
Favoretto CA, Righi T, Fernandes GJD, Bertagna NB, Rodolpho BT, Janisset NDRLDL, Jovita-Farias C, Costa GVL, Anjos-Santos AD, Romualdo da Silva FB, Leão RM, Cruz FC. Animal models for studying therapeutic targets and treatments for alcohol use disorder. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 178:355-381. [PMID: 39523060 DOI: 10.1016/bs.irn.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Over the decades, preclinical models have been developed and refined to investigate the rewarding effects of addictive substances and the neurobiological underpinnings of alcohol and other drug use disorders. This chapter delves into the methodological foundations, advantages, and limitations of leading animal models used to study alcohol use disorders (AUDs). Some models focus on the early stages of alcohol use and abuse. For instance, conditioned place preference assesses associative learning between a specific context and the effects of the drug, while locomotor sensitization measures increased locomotor activity following repeated drug exposure. In contrast, contingent models such as operant and non-operant alcohol self-administration protocols gauge voluntary intake, preference, motivation, and seeking behavior for alcohol solutions among experimental subjects. Additionally, we discuss the chronic intermittent alcohol vapor model, extensively utilized to induce a phenotype resembling dependence through non-contingent inhalation of alcohol vapor, resulting in elevated blood alcohol concentrations. Given the focus on pharmacological treatments for AUDs, we explore how different animal models can be employed to evaluate potential therapies and extrapolate findings to alcohol-related behaviors in humans. This chapter aims to provide readers with a comprehensive understanding of various animal models for AUDs, aiding in the interpretation of preclinical studies and the selection of suitable models for future research endeavors.
Collapse
Affiliation(s)
- Cristiane Aparecida Favoretto
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Thamires Righi
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Gustavo Juliate Damaceno Fernandes
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Natalia Bonetti Bertagna
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Ben Tagami Rodolpho
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Nilma do Rocio Lara de Lima Janisset
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Caio Jovita-Farias
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Giovanna Victória Lopes Costa
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Alexia Dos Anjos-Santos
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | | | - Rodrigo Molini Leão
- Laboratory of Pharmacology, Biomedical Sciences Institute, Department of Pharmacology, Federal University of Uberlândia, Uberlândia, MG, Brazil
| | - Fábio Cardoso Cruz
- Molecular and Behavioral Neuroscience Laboratory, Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil.
| |
Collapse
|
23
|
Lopez S, Rufener C, Holinger M. How do different amounts of straw as well as compost in the home pen affect the rooting motivation of growing-finishing pigs? Animal 2024; 18:101325. [PMID: 39357491 DOI: 10.1016/j.animal.2024.101325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Rooting is a strongly motivated, species-specific behaviour of pigs. Most housing systems do not provide appropriate materials that enable the full expression of this behaviour, and it remains unclear whether straw is suitable to entirely fulfil the rooting motivation of pigs. We therefore investigated the suitability of small (minimal) and large (deep) amounts of straw as well as large amounts of compost to satisfy rooting motivation in pigs. Fifty-seven growing-finishing pigs were housed in three pens, each providing permanent access to one of the three treatment substrates. Eight pigs per group were tested individually in a classical preference test (PT) and another eight pigs in a conditioned place preference test (CPPT). In the tests, pigs could show their preference to consume freely available feed ("feed") or feed hidden in sawdust ("root"). In the CPPT, feed was only present during training but not during testing. Pigs were exposed to the test situation twice, with approximately 72 kg and 115 kg BW. In both tests, the following variables were measured and used as outcome variables in linear mixed effect models: first decision to choose one of the two stimuli ("feed" or "root"), duration of time spent in proximity to "root", number of changes between stimuli, and latency to the first decision. Overall, the pigs' first decision (by tendency; P = 0.076) and the duration in proximity to "root" (P = 0.034) varied among treatments: Pigs housed with minimal straw tended to be more likely to choose "root" first (posthoc comparison; P = 0.090) and spent more time in proximity to "root" (P = 0.030) than pigs housed with compost, whereas pigs housed with deep straw were intermediate. Interestingly, the patterns of response to the treatment differed depending on the behavioural tests for both, first decision (interaction; P = 0.032) and duration in proximity to "root" (interaction; by tendency; P = 0.006). In addition, pigs in the PT changed more often between stimuli than pigs in the CPPT (P < 0.001). There was a tendency for an interactive effect between test and treatment for latency to first decision (interaction; P = 0.082), though pairwise comparisons did not reveal any differences. We concluded that in this study housing with permanent access to compost satisfied rooting motivation in pigs more than housing with minimal amounts of straw.
Collapse
Affiliation(s)
- S Lopez
- Centre for Proper Housing of Ruminants and Pigs, Federal Food Safety and Veterinary Office, Agroscope, 8356 Ettenhausen, Switzerland; University of Bern, Veterinary Public Health Institute, Vetsuisse Faculty, 3012 Bern, Switzerland
| | - C Rufener
- Centre for Proper Housing of Ruminants and Pigs, Federal Food Safety and Veterinary Office, Agroscope, 8356 Ettenhausen, Switzerland.
| | - M Holinger
- Research Institute of Organic Agriculture (FiBL), Department of Livestock Sciences, 5070 Frick, Switzerland
| |
Collapse
|
24
|
Ide S, Ikeda K. Caenorhabditis elegans for opioid addiction research. Curr Opin Neurobiol 2024; 88:102914. [PMID: 39236640 DOI: 10.1016/j.conb.2024.102914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 07/16/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
The problem of drug addiction has become a profound societal problem worldwide. A better understanding of the neurobiological basis of addiction and the discovery of more effective treatments are needed. Recent studies have shown that many mechanisms that underlie addiction exist in more primitive organisms, including the nematode Caenorhabditis elegans (C. elegans). C. elegans is also hypothesized to possess a functional opioid-like system, including the endogenous opioid-like peptide NLP-24 and opioid-like receptor NPR-17. Opioids, such as morphine, are thought to cause addiction-like behavior by activating dopamine nerves in C. elegans via the opioid-like system. Accumulating evidence suggests that C. elegans is an excellent animal model for identifying molecular mechanisms of addiction.
Collapse
Affiliation(s)
- Soichiro Ide
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan.
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-Ku, Tokyo, Japan; Department of Neuropsychopharmacology, National Institute of Mental Health, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi-cho, Kodaira, Tokyo, Japan
| |
Collapse
|
25
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [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: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
26
|
Vanderhoof SO, Vincent CJ, Beaver JN, Latsko MS, Aguilar-Alvarez R, Jasnow AM. Corticosterone after early adolescent stress prevents social avoidance, aversive behavior, and morphine-conditioned place preference in adulthood. Psychopharmacology (Berl) 2024; 241:2045-2059. [PMID: 38805040 PMCID: PMC11442498 DOI: 10.1007/s00213-024-06616-7] [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: 01/08/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
RATIONALE Stress during childhood or adolescence increases vulnerability to psychiatric disorders in adults. In adult rodents, the delayed effects of stress can increase anxiety-like behavior. These effects, however, can be prevented with post-stress administration of corticosterone (CORT). The effectiveness of CORT in preventing adolescent stress-induced emotional behavior alterations in adulthood has yet to be investigated. OBJECTIVES Here, we investigated the interactions between early adolescent stress and exogenous corticosterone on adult social, aversive, and drug-seeking behavior in mice, which are translationally related to symptoms associated with psychiatric and substance abuse disorders. METHODS AND RESULTS A single administration of CORT in drinking water (400ug/mL) for 24 h after social defeat or context fear conditioning prevents defeat-induced social avoidance, alters fear processing, prevents adolescent stress-induced anhedonia, and prevents stress-potentiated morphine place preference in adulthood. Exogenous CORT did not immediately prevent stress-induced potentiation of morphine conditioned-place preference in adolescents but did so in adult mice. However, when administered to adolescent mice, CORT also prevented the incubation of morphine-conditioned place preference into adulthood. Lastly, exogenous CORT administration blunted endogenous corticosterone but was unrelated to freezing behavior during a fear test. CONCLUSIONS This is the first demonstration of adolescent post-stress CORT promoting socio-emotional resilience and preventing drug-seeking behavior. Our data suggest elevated corticosterone after a stress experience promotes resilience for at least 40 days across the developmental transition from adolescence to adulthood and is effective for socio-emotional and drug-seeking behavior. These results are critical for understanding how adolescent stress impacts emotional and drug-seeking behavior into adulthood.
Collapse
Affiliation(s)
- Samantha O Vanderhoof
- Department of Psychological Sciences, Brain Health Research Institute, Kent State University, Kent, USA
| | - Carly J Vincent
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, USA
| | - Jasmin N Beaver
- Department of Psychological Sciences, Brain Health Research Institute, Kent State University, Kent, USA
| | - Maeson S Latsko
- Department of Psychological Sciences, Brain Health Research Institute, Kent State University, Kent, USA
| | - Ricardo Aguilar-Alvarez
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, USA
| | - Aaron M Jasnow
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, USA.
| |
Collapse
|
27
|
Kim OH, Jeon KO, Kim G, Jang CG, Yoon SS, Jang EY. The neuropharmacological properties of α-pyrrolidinobutiothiophenone, a new synthetic cathinone, in rodents; role of the dopaminergic system. Br J Pharmacol 2024; 181:3462-3482. [PMID: 38772548 DOI: 10.1111/bph.16422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/14/2024] [Accepted: 04/19/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND AND PURPOSE α-Pyrrolidinobutiothiophenone (α-PBT) is a chemical derivative of cathinone, a structural analogue of amphetamine. Until now, there have been a few previous neurochemical or neurobehavioural studies on the abuse potential of α-PBT. EXPERIMENTAL APPROACH We examined the abuse potential of α-PBT by measuring psychomotor, rewarding, and reinforcing properties and methamphetamine-like discriminative stimulus effects in rodents using locomotor activity, conditioned place preference, self-administration, and drug discrimination studies. To clarify the underlying neuropharmacological mechanisms, we measured dopamine levels and neuronal activation in the dorsal striatum. In addition, we investigated the role of the dopamine D1 receptor or D2 receptors in α-PBT-induced hyperlocomotor activity, conditioned place preference, and the methamphetamine-like discriminative stimulus effect of α-PBT in rodents. KEY RESULTS α-PBT promoted hyperlocomotor activity in mice. α-PBT induced drug-paired place preference in mice and supported self-administration in rats. In a drug discrimination experiment, α-PBT fully substituted for the discriminative stimulus effects of methamphetamine in rats. Furthermore, α-PBT increased dopamine levels and c-Fos expression in the dorsal striatum of mice, which was associated with these behaviours. Finally, pretreatment with the D1 receptor antagonist SCH23390 or the D2 receptors antagonist eticlopride significantly attenuated acute or repeated α-PBT-induced hyperlocomotor activity, place preference, and the methamphetamine-like discriminative stimulus effects in rodents. CONCLUSIONS AND IMPLICATIONS These findings suggest that α-PBT has abuse potential at the highest dose tested via enhanced dopaminergic transmission in the dorsal striatum of rodents. The results provide scientific evidence for the legal restrictions of the recreational use of α-PBT.
Collapse
Affiliation(s)
- Oc-Hee Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Kyung Oh Jeon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Gihyeon Kim
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seong Shoon Yoon
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, Republic of Korea
| | - Eun Young Jang
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Kim YJ, Kim K, Lee Y, Min HW, Ko YH, Lee BR, Hur KH, Kim SK, Lee SY, Jang CG. The mutated cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2 S968F) regulates cocaine-induced reward behaviour and plasticity in the nucleus accumbens. Br J Pharmacol 2024; 181:3327-3345. [PMID: 38751203 DOI: 10.1111/bph.16427] [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: 12/21/2023] [Revised: 04/02/2024] [Accepted: 04/25/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND AND PURPOSE Cytoplasmic fragile X messenger ribonucleoprotein 1 (FMR1)-interacting protein 2 (CYFIP2), as a component of the Wiskott-Aldrich syndrome protein family verprolin-homologous protein (WAVE) regulatory complex, is involved in actin polymerization, contributing to neuronal development and structural plasticity. Mutating serine-968 to phenylalanine (S968F) in CYFIP2 causes an altered cocaine response in mice. The neuronal mechanisms underlying this response remain unknown. EXPERIMENTAL APPROACH We performed cocaine reward-related behavioural tests and examined changes in synaptic protein phenotypes and neuronal morphology in the nucleus accumbens (NAc), using CYFIP2 S968F knock-in mice to investigate the role of CYFIP2 in regulating cocaine reward. KEY RESULTS CYFIP2 S968F mutation attenuated cocaine-induced behavioural sensitization and conditioned place preference. Cocaine-induced c-Fos was not observed in the NAc of CYFIP2 S968F knock-in mice. However, c-Fos induction was still evident in the medial prefrontal cortex (mPFC). CYFIP2 S968F mutation altered cocaine-associated CYFIP2 signalling, glutamatergic protein expression and synaptic density in the NAc following cocaine exposure. To further determine the role of CYFIP2 in NAc neuronal activity and the mPFC projecting to the NAc activity-mediating reward response, we used optogenetic tools to stimulate the NAc or mPFC-NAc pathway and observed that optogenetic activation of the NAc or mPFC-NAc pathway induced reward-related behaviours. This effect was not observed in the S968F mutation in CYFIP2. CONCLUSION AND IMPLICATIONS These results suggest that CYFIP2 plays a role in controlling cocaine-mediated neuronal function and structural plasticity in the NAc, and that CYFIP2 could serve as a target for regulating cocaine reward.
Collapse
Affiliation(s)
- Young-Jung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kyungin Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hee-Won Min
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yong-Hyun Ko
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
29
|
Yates JR. Pharmacological Treatments for Methamphetamine Use Disorder: Current Status and Future Targets. Subst Abuse Rehabil 2024; 15:125-161. [PMID: 39228432 PMCID: PMC11370775 DOI: 10.2147/sar.s431273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
The illicit use of the psychostimulant methamphetamine (METH) is a major concern, with overdose deaths increasing substantially since the mid-2010s. One challenge to treating METH use disorder (MUD), as with other psychostimulant use disorders, is that there are no available pharmacotherapies that can reduce cravings and help individuals achieve abstinence. The purpose of the current review is to discuss the molecular targets that have been tested in assays measuring the physiological, the cognitive, and the reinforcing effects of METH in both animals and humans. Several drugs show promise as potential pharmacotherapies for MUD when tested in animals, but fail to produce long-term changes in METH use in dependent individuals (eg, modafinil, antipsychotic medications, baclofen). However, these drugs, plus medications like atomoxetine and varenicline, may be better served as treatments to ameliorate the psychotomimetic effects of METH or to reverse METH-induced cognitive deficits. Preclinical studies show that vesicular monoamine transporter 2 inhibitors, metabotropic glutamate receptor ligands, and trace amine-associated receptor agonists are efficacious in attenuating the reinforcing effects of METH; however, clinical studies are needed to determine if these drugs effectively treat MUD. In addition to screening these compounds in individuals with MUD, potential future directions include increased emphasis on sex differences in preclinical studies and utilization of pharmacogenetic approaches to determine if genetic variances are predictive of treatment outcomes. These future directions can help lead to better interventions for treating MUD.
Collapse
Affiliation(s)
- Justin R Yates
- Department of Psychological Science, Northern Kentucky University, Highland Heights, KY, USA
| |
Collapse
|
30
|
Hur KH, Lee Y, Donio AL, Kim SK, Lee BR, Seo JY, Kundu D, Kim KM, Kohut SJ, Lee SY, Jang CG. Transient receptor potential ankyrin 1 channel modulates the abuse-related mechanisms of methamphetamine through interaction with dopamine transporter. Br J Pharmacol 2024; 181:2794-2809. [PMID: 38644533 PMCID: PMC11230846 DOI: 10.1111/bph.16370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/17/2024] [Accepted: 02/29/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND AND PURPOSE Methamphetamine (METH) use disorder has risen dramatically over the past decade, and there are currently no FDA-approved medications due, in part, to gaps in our understanding of the pharmacological mechanisms related to METH action in the brain. EXPERIMENTAL APPROACH Here, we investigated whether transient receptor potential ankyrin 1 (TRPA1) mediates each of several METH abuse-related behaviours in rodents: self-administration, drug-primed reinstatement, acquisition of conditioned place preference, and hyperlocomotion. Additionally, METH-induced molecular (i.e., neurotransmitter and protein) changes in the brain were compared between wild-type and TRPA1 knock-out mice. Finally, the relationship between TRPA1 and the dopamine transporter was investigated through immunoprecipitation and dopamine reuptake assays. KEY RESULTS TRPA1 antagonism blunted METH self-administration and drug-primed reinstatement of METH-seeking behaviour. Further, development of METH-induced conditioned place preference and hyperlocomotion were inhibited by TRPA1 antagonist treatment, effects that were not observed in TRPA1 knock-out mice. Similarly, molecular studies revealed METH-induced increases in dopamine levels and expression of dopamine system-related proteins in wild-type, but not in TRPA1 knock-out mice. Furthermore, pharmacological blockade of TRPA1 receptors reduced the interaction between TRPA1 and the dopamine transporter, thereby increasing dopamine reuptake activity by the transporter. CONCLUSION AND IMPLICATIONS This study demonstrates that TRPA1 is involved in the abuse-related behavioural effects of METH, potentially through its modulatory role in METH-induced activation of dopaminergic neurotransmission. Taken together, these data suggest that TRPA1 may be a novel therapeutic target for treating METH use disorder.
Collapse
Affiliation(s)
- Kwang-Hyun Hur
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Behavioral Neuroimaging Laboratory, McLean Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Youyoung Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Audrey Lynn Donio
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Seon-Kyung Kim
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Bo-Ram Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jee-Yeon Seo
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Dooti Kundu
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju, Republic of Korea
| | - Stephen J Kohut
- Behavioral Neuroimaging Laboratory, McLean Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Seok-Yong Lee
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
31
|
Mozafari R, Khodagholi F, Kaveh N, Zibaii ME, Kalivas P, Haghparast A. Blockade of mGluR5 in nucleus accumbens modulates calcium sensor proteins, facilitates extinction, and attenuates reinstated morphine place preference in rats. J Psychiatr Res 2024; 176:23-32. [PMID: 38833749 DOI: 10.1016/j.jpsychires.2024.05.047] [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: 01/11/2024] [Revised: 05/02/2024] [Accepted: 05/29/2024] [Indexed: 06/06/2024]
Abstract
Numerous findings confirm that the metabotropic glutamate receptors (mGluRs) are involved in the conditioned place preference (CPP) induced by morphine. Here we focused on the role of mGluR5 in the nucleus accumbens (NAc) as a main site of glutamate action on the rewarding effects of morphine. Firstly, we investigated the effects of intra-NAc administrating mGluR5 antagonist 3-((2-Methyl-1,3-thiazol-4-yl) ethynyl) pyridine hydrochloride (MTEP; 1, 3, and 10 μg/μl saline) on the extinction and the reinstatement phase of morphine CPP. Moreover, to determine the downstream signaling cascades of mGluR5 in morphine CPP, the protein levels of stromal interaction molecules (STIM1 and 2) in the NAc and hippocampus (HPC) were measured by western blotting. The behavioral data indicated that the mGluR5 blockade by MTEP at the high doses of 3 and 10 μg facilitated the extinction of morphine-induced CPP and attenuated the reinstatement to morphine in extinguished rats. Molecular results showed that the morphine led to increased levels of STIM proteins in the HPC and increased the level of STIM1 without affecting STIM2 in the NAc. Furthermore, intra-NAc microinjection of MTEP (10 μg) in the reinstatement phase decreased STIM1 in the NAc and HPC and reduced the STIM2 in the HPC. Collectively, our data show that morphine could facilitate brain reward function in part by increasing glutamate-mediated transmission through activation of mGluR5 and modulation of STIM proteins. Therefore, these results highlight the therapeutic potential of mGluR5 antagonists in morphine use disorder.
Collapse
Affiliation(s)
- Roghayeh Mozafari
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Kaveh
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Peter Kalivas
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran; Department of Basic Sciences, Iranian Academy of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Cuttoli RDD, Issler O, Yakubov B, Jahan N, Abid A, Kasparov S, Granizo K, Ahmed S, Russo SJ, Nestler EJ, Sweis BM. Sex differences in change-of-mind neuroeconomic decision-making is modulated by LINC00473 in medial prefrontal cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.592609. [PMID: 39005412 PMCID: PMC11244910 DOI: 10.1101/2024.05.08.592609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Changing one's mind is a complex cognitive phenomenon involving a continuous re-appraisal of the trade-off between past costs and future value. Recent work modeling this behavior across species has established associations between aspects of this choice process and their contributions to altered decision-making in psychopathology. Here, we investigated the actions in medial prefrontal cortex (mPFC) neurons of long intergenic non-coding RNA, LINC00473, known to induce stress resilience in a striking sex-dependent manner, but whose role in cognitive function is unknown. We characterized complex decision-making behavior in male and female mice longitudinally in our neuroeconomic foraging paradigm, Restaurant Row, following virus-mediated LINC00473 expression in mPFC neurons. On this task, mice foraged for their primary source of food among varying costs (delays) and subjective value (flavors) while on a limited time-budget during which decisions to accept and wait for rewards were separated into discrete stages of primary commitments and secondary re-evaluations. We discovered important differences in decision-making behavior between female and male mice. LINC00473 expression selectively influenced multiple features of re-evaluative choices, without affecting primary decisions, in female mice only. These behavioral effects included changing how mice (i) cached the value of the passage of time and (ii) weighed their history of economically disadvantageous choices. Both processes were uniquely linked to change-of-mind decisions and underlie the computational bases of distinct aspects of counterfactual thinking. These findings reveal a key bridge between a molecular driver of stress resilience and psychological mechanisms underlying sex-specific decision-making proclivities.
Collapse
|
33
|
Schwarting RKW, Wöhr M, Engler H, Sungur AÖ, Schedlowski M. Behaviorally conditioned effects of psychoactive drugs in experimental animals: What we have learned from nearly a century of research and what remains to be learned. Neurosci Biobehav Rev 2024; 162:105721. [PMID: 38754716 DOI: 10.1016/j.neubiorev.2024.105721] [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: 03/09/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Continuous treatment with drugs is a crucial requirement for managing various clinical conditions, including chronic pain and neuropsychiatric disorders such as depression or schizophrenia. Associative learning processes, i.e. Pavlovian conditioning, can play an important role for the effects of drugs and could open new avenues for optimizing patient treatment. In this narrative literature review, we summarize available data in experimental animals regarding the behaviorally conditioned effects of psychostimulants such as d-amphetamine and cocaine, the dopamine receptor agonist apomorphine, the dopamine receptor antagonist haloperidol, morphine and antidepressant drugs. In each section, the drug under discussion is briefly introduced, followed by a detailed examination of conditioning features, including doses and dosing regimens, characteristics of the conditioning process such as test environments or specific conditioned stimuli, testing and conditioned response characteristics, possible extinction or reconditioning or reversal training, neural mechanisms, and finally, the potential clinical relevance of the research area related to the drug. We focus on key outcomes, delve into methodical issues, identify gaps in current knowledge, and suggest future research directions.
Collapse
Affiliation(s)
- Rainer K W Schwarting
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany
| | - Markus Wöhr
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany; KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Leuven B-3000, Belgium; KU Leuven, Leuven Brain Institute, Leuven B-3000, Belgium
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro-, and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen D-45147, Germany
| | - A Özge Sungur
- Philipps-University of Marburg, Faculty of Psychology, Experimental and Biological Psychology, Behavioral Neuroscience, Marburg D-35032, Germany; Center for Mind, Brain and Behavior, Marburg D-35032, Germany; KU Leuven, Faculty of Psychology and Educational Sciences, Research Unit Brain and Cognition, Laboratory of Biological Psychology, Social and Affective Neuroscience Research Group, Leuven B-3000, Belgium; KU Leuven, Leuven Brain Institute, Leuven B-3000, Belgium
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, Center for Translational Neuro-, and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen D-45147, Germany; Department of Clinical Neuroscience, Osher Center for Integrative Medicine, Karolinska Institutet, Stockholm 171 77, Sweden
| |
Collapse
|
34
|
Socha J, Grochecki P, Marszalek-Grabska M, Skrok A, Smaga I, Slowik T, Prazmo W, Kotlinski R, Filip M, Kotlinska JH. Cannabidiol Protects against the Reinstatement of Oxycodone-Induced Conditioned Place Preference in Adolescent Male but Not Female Rats: The Role of MOR and CB1R. Int J Mol Sci 2024; 25:6651. [PMID: 38928357 PMCID: PMC11204276 DOI: 10.3390/ijms25126651] [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: 04/11/2024] [Revised: 06/04/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Cannabidiol (CBD), a phytocannabinoid, appeared to satisfy several criteria for a safe approach to preventing drug-taking behavior, including opioids. However, most successful preclinical and clinical results come from studies in adult males. We examined whether systemic injections of CBD (10 mg/kg, i.p.) during extinction of oxycodone (OXY, 3 mg/kg, i.p.) induced conditioned place preference (CPP) could attenuate the reinstatement of CPP brought about by OXY (1.5 mg/kg, i.p.) priming in adolescent rats of both sexes, and whether this effect is sex dependent. Accordingly, a priming dose of OXY produced reinstatement of the previously extinguished CPP in males and females. In both sexes, this effect was linked to locomotor sensitization that was blunted by CBD pretreatments. However, CBD was able to prevent the reinstatement of OXY-induced CPP only in adolescent males and this outcome was associated with an increased cannabinoid 1 receptor (CB1R) and a decreased mu opioid receptor (MOR) expression in the prefrontal cortex (PFC). The reinstatement of CCP in females was associated with a decreased MOR expression, but no changes were detected in CB1R in the hippocampus (HIP). Moreover, CBD administration during extinction significantly potentialized the reduced MOR expression in the PFC of males and showed a tendency to potentiate the reduced MOR in the HIP of females. Additionally, CBD reversed OXY-induced deficits of recognition memory only in males. These results suggest that CBD could reduce reinstatement to OXY seeking after a period of abstinence in adolescent male but not female rats. However, more investigation is required.
Collapse
Affiliation(s)
- Justyna Socha
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Pawel Grochecki
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Marta Marszalek-Grabska
- Department of Experimental and Clinical Pharmacology, Medical University, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Aleksandra Skrok
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| | - Irena Smaga
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (M.F.)
| | - Tymoteusz Slowik
- Experimental Medicine Center, Medical University, Jaczewskiego 8, 20-090 Lublin, Poland;
| | - Wojciech Prazmo
- Breast Surgery Department, Provincial Specialist Hospital, Al. Krasnicka 100, 20-718 Lublin, Poland;
| | - Robert Kotlinski
- Clinical Department of Cardiac Surgery, University of Rzeszow, Lwowska 60, 35-301 Rzeszow, Poland;
| | - Malgorzata Filip
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Smetna 12, 31-343 Krakow, Poland; (I.S.); (M.F.)
| | - Jolanta H. Kotlinska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland; (J.S.); (P.G.); (A.S.)
| |
Collapse
|
35
|
Ahmadianmoghadam MA, Nematollahi MH, Mehrabani M, Fatemi I, Rostamzadeh F, Dell'Agli M, Mehrabani M, Abolhassani M, Mehrbani M. Effect of an herbal formulation containing Peganum harmala L. and Fraxinus excelsior L. on oxidative stress, memory impairment and withdrawal syndrome induced by morphine. Int J Neurosci 2024; 134:570-583. [PMID: 36168934 DOI: 10.1080/00207454.2022.2130293] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 09/05/2022] [Accepted: 09/15/2022] [Indexed: 10/17/2022]
Abstract
Background: Traditional Persian medicine has introduced effective remedies in opioid dependence care. One of the most widely used remedies is an herbal formulation containing Peganum harmala L. and Fraxinus excelsior L. (HF). This study investigated the effects of HF to attenuate the withdrawal signs and rewarding effects in morphine-dependent rats. Methods: Forty-nine male Wistar rats were randomly divided into seven groups. The control and vehicle groups received normal saline and sodium carboxymethyl cellulose, respectively. The morphine group received morphine for one week. The single and daily dose of HF groups received morphine similar to the morphine group, and HF (1.4 and 2.8 g/kg) once a day in the daily dose group and only on the last day of the experiment in the single dose of HF group. Finally, the withdrawal signs as well biochemical tests were evaluated. The behavioral parameters were assessed by conditioned place preference (CPP), elevated plus-maze and Y-maze tests. The antioxidant activity of HF was evaluated by measurement of serum contents of malondialdehyde, stable nitric oxide metabolites and total antioxidant capacity (TAC). Moreover, the protein expression of c-fos was assessed by western blotting. Results: Daily treatment with HF significantly reduced the score of CPP behavioral test, all of the withdrawal signs, TAC and the c-fos protein level. Conclusions: The results indicated that HF might be a promising complementary treatment in reducing morphine-induced physical and psychological dependence probably through modulation of c-fos protein expression.
Collapse
Affiliation(s)
- Mohammad Ali Ahmadianmoghadam
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Department of Traditional Medicine, Faculty of Persian Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Hadi Nematollahi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Mehrabani
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Iman Fatemi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mario Dell'Agli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Moslem Abolhassani
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehrzad Mehrbani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
36
|
Monari PK, Hammond ER, Zhao X, Maksimoski AN, Petric R, Malone CL, Riters LV, Marler CA. Conditioned preferences: Gated by experience, context, and endocrine systems. Horm Behav 2024; 161:105529. [PMID: 38492501 DOI: 10.1016/j.yhbeh.2024.105529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024]
Abstract
Central to the navigation of an ever-changing environment is the ability to form positive associations with places and conspecifics. The functions of location and social conditioned preferences are often studied independently, limiting our understanding of their interplay. Furthermore, a de-emphasis on natural functions of conditioned preferences has led to neurobiological interpretations separated from ecological context. By adopting a naturalistic and ethological perspective, we uncover complexities underlying the expression of conditioned preferences. Development of conditioned preferences is a combination of motivation, reward, associative learning, and context, including for social and spatial environments. Both social- and location-dependent reward-responsive behaviors and their conditioning rely on internal state-gating mechanisms that include neuroendocrine and hormone systems such as opioids, dopamine, testosterone, estradiol, and oxytocin. Such reinforced behavior emerges from mechanisms integrating past experience and current social and environmental conditions. Moreover, social context, environmental stimuli, and internal state gate and modulate motivation and learning via associative reward, shaping the conditioning process. We highlight research incorporating these concepts, focusing on the integration of social neuroendocrine mechanisms and behavioral conditioning. We explore three paradigms: 1) conditioned place preference, 2) conditioned social preference, and 3) social conditioned place preference. We highlight nonclassical species to emphasize the naturalistic applications of these conditioned preferences. To fully appreciate the complex integration of spatial and social information, future research must identify neural networks where endocrine systems exert influence on such behaviors. Such research promises to provide valuable insights into conditioned preferences within a broader naturalistic context.
Collapse
Affiliation(s)
- Patrick K Monari
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA.
| | - Emma R Hammond
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA
| | - Xin Zhao
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA
| | - Alyse N Maksimoski
- University of Wisconsin-Madison, Department of Integrative Biology, Madison, WI, USA
| | - Radmila Petric
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA; Institute for the Environment, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Candice L Malone
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA
| | - Lauren V Riters
- University of Wisconsin-Madison, Department of Integrative Biology, Madison, WI, USA
| | - Catherine A Marler
- University of Wisconsin-Madison, Department of Psychology, Madison, WI, USA; University of Wisconsin-Madison, Department of Integrative Biology, Madison, WI, USA.
| |
Collapse
|
37
|
Pastor V, Medina JH. α7 nicotinic acetylcholine receptor in memory processing. Eur J Neurosci 2024; 59:2138-2154. [PMID: 36634032 DOI: 10.1111/ejn.15913] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
Information storage in the brain involves different memory types and stages that are processed by several brain regions. Cholinergic pathways through acetylcholine receptors actively participate on memory modulation, and their disfunction is associated with cognitive decline in several neurological disorders. During the last decade, the role of α7 subtype of nicotinic acetylcholine receptors in different memory stages has been studied. However, the information about their role in memory processing is still scarce. In this review, we attempt to identify brain areas where α7 nicotinic receptors have an essential role in different memory types and stages. In addition, we discuss recent work implicating-or not-α7 nicotinic receptors as promising pharmacological targets for memory impairment associated with neurological disorders.
Collapse
Affiliation(s)
- Verónica Pastor
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Facultad de Medicina, Departamento de Ciencias Fisiológicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge H Medina
- Instituto de Biología Celular y Neurociencia "Prof. Eduardo De Robertis" (IBCN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto Tecnológico de Buenos Aires (ITBA), Buenos Aires, Argentina
| |
Collapse
|
38
|
Jeon KO, Kim OH, Seo SY, Yun J, Jang CG, Lim RN, Kim TW, Yang CH, Yoon SS, Jang EY. The psychomotor, reinforcing, and discriminative stimulus effects of synthetic cathinone mexedrone in male mice and rats. Eur J Pharmacol 2024; 969:176466. [PMID: 38431243 DOI: 10.1016/j.ejphar.2024.176466] [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: 01/22/2024] [Revised: 02/18/2024] [Accepted: 02/28/2024] [Indexed: 03/05/2024]
Abstract
The chronic use of the novel synthetic cathinone mexedrone, like other psychoactive drugs, can be considered addictive, with a high potential for abuse and the ability to cause psychological dependence in certain users. However, little is known about the neurobehavioral effects of mexedrone in association with its potential for abuse. We investigated the abuse potential for mexedrone abuse through multiple behavioral tests. In addition, serotonin transporter (SERT) levels were measured in the synaptosome of the dorsal striatum, and serotonin (5-HT) levels were measured in the dorsal striatum of acute mexedreone (50 mg/kg)-treated mice. To clarify the neuropharmacological mechanisms underlying the locomotor response of mexedrone, the 5-HT2A receptor antagonist M100907 (0.5 or 1.0 mg/kg) was administered prior to the acute injection of mexedrone in the locomotor activity experiment in mice. Mexedrone (10-50 mg/kg) produced a significant place preference in mice and mexedrone (0.1-0.5 mg/kg/infusion) maintained self-administration behavior in rats in a dose-dependent manner. In the drug discrimination experiment, mexedrone (5.6-32 mg/kg) was fully substituted for the discriminative stimulus effects of cocaine in rats. Mexedrone increased locomotor activity, and these effects were reversed by pretreatment with M100907. Acute mexedrone significantly increased c-Fos expression in the dorsal striatum and decreased SERT levels in the synaptosome of the dorsal striatum of mice, resulting in an elevation of 5-HT levels. Taken together, our results provide the possibility that mexedrone has abuse potential, which might be mediated, at least in part, by the activation of the serotonergic system in the dorsal striatum.
Collapse
Affiliation(s)
- Kyung Oh Jeon
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea; Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Oc-Hee Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Su Yeon Seo
- Korean Medicine (KM) Research Division, Korea Institute of Oriental Medicine, Daejeon, 34054, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy, Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do, 28160, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Ri-Na Lim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Tae Wan Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Chae Ha Yang
- Department of Physiology, College of Korean Medicine, Daegu Haany University, 136 Sincheondong-ro, Suseong-gu, Daegu, 42158, Republic of Korea
| | - Seong Shoon Yoon
- Department of Physiology, College of Korean Medicine, Daegu Haany University, 136 Sincheondong-ro, Suseong-gu, Daegu, 42158, Republic of Korea.
| | - Eun Young Jang
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
39
|
Martínez-Rivera A, Fetcho RN, Birmingham L, Jiu JX, Yang R, Foord C, Scala-Chávez D, Mekawy N, Pleil K, Pickel VM, Liston C, Castorena CM, Levitz J, Pan YX, Briand LA, Rajadhyaksha AM, Lee FS. Elevating levels of the endocannabinoid 2-arachidonoylglycerol blunts opioid reward but not analgesia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.585967. [PMID: 38766079 PMCID: PMC11101127 DOI: 10.1101/2024.04.02.585967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Converging findings have established that the endocannabinoid (eCB) system serves as a possible target for the development of new treatments for pain as a complement to opioid-based treatments. Here we show in male and female mice that enhancing levels of the eCB, 2-arachidonoylglycerol (2-AG), through pharmacological inhibition of its catabolic enzyme, monoacylglycerol lipase (MAGL), either systemically or in the ventral tegmental area (VTA) with JZL184, leads to a substantial attenuation of the rewarding effects of opioids in male and female mice using conditioned place preference and self-administration paradigms, without altering their analgesic properties. These effects are driven by CB1 receptors (CB1Rs) within the VTA as VTA CB1R conditional knockout, counteracts JZL184's effects. Conversely, pharmacologically enhancing the levels of the other eCB, anandamide (AEA), by inhibition of fatty acid amide hydrolase (FAAH) has no effect on opioid reward or analgesia. Using fiber photometry with fluorescent sensors for calcium and dopamine (DA), we find that enhancing 2-AG levels diminishes opioid reward-related nucleus accumbens (NAc) activity and DA neurotransmission. Together these findings reveal that 2-AG counteracts the rewarding properties of opioids and provides a potential adjunctive therapeutic strategy for opioid-related analgesic treatments.
Collapse
Affiliation(s)
- Arlene Martínez-Rivera
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Robert N. Fetcho
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lizzie Birmingham
- Department of Psychology, Temple University; Neuroscience Program, Temple University, 19122, USA
| | - Jin X Jiu
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Ruirong Yang
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Careen Foord
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Diego Scala-Chávez
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Narmin Mekawy
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Kristen Pleil
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Virginia M. Pickel
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Conor Liston
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carlos M. Castorena
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joshua Levitz
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ying-Xian Pan
- Department of Anesthesiology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Lisa A. Briand
- Department of Psychology, Temple University; Neuroscience Program, Temple University, 19122, USA
| | - Anjali M. Rajadhyaksha
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA, USA
- Division of Pediatric Neurology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Francis S. Lee
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
40
|
Keller BN, Snyder AE, Coker CR, Aguilar EA, O’Brien MK, Bingaman SS, Arnold AC, Hajnal A, Silberman Y. Vagus nerve damage increases alcohol intake and preference in a nonpreferring rat line: Relationship to vagal regulation of the hypothalamic-pituitary-adrenal axis. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:488-498. [PMID: 38311347 PMCID: PMC10939901 DOI: 10.1111/acer.15264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/12/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024]
Abstract
BACKGROUND Clinical and preclinical research indicates that gastric weight loss surgeries, such as Roux-en-Y gastric bypass surgery, can induce alcohol use disorder (AUD). While numerous mechanisms have been proposed for these effects, one relatively unexplored potential mechanism is physical damage to the gastric branch of the vagus nerve, which can occur during bypass surgery. Therefore, we hypothesized that direct damage to the gastric branch of the vagus nerve, without altering other aspects of gastric anatomy, could result in increased alcohol intake. METHODS To test this hypothesis, we compared alcohol intake and preference in multiple models in male Sprague-Dawley rats that received selective gastric branch vagotomy (VX) with rats who underwent sham surgery. Because the vagus nerve regulates hypothalamic-pituitary-adrenal (HPA) axis function, and alterations to HPA function are critical to the escalation of non-dependent alcohol intake, we also tested the hypothesis that gastric VX increases HPA function. RESULTS We found that VX increases alcohol intake and preference in the every-other-day, two-bottle choice test and increases preference for 1 g/kg alcohol in the conditioned place preference test. The effects were selective for alcohol, as sucrose intake and preference were not altered by VX. We also found that VX increases corticotropin releasing factor (CRF) mRNA in the paraventricular nucleus of the hypothalamus (PVN), increases putative PVN CRF neuronal action potential firing, and increases corticosterone levels. CONCLUSIONS Overall, these findings suggest that the vagus nerve may play a critical role in regulating HPA axis function via modulation of PVN CRF mRNA expression and putative PVN CRF neuronal activity. Furthermore, disruptions to vagal regulation of HPA axis function may increase alcohol intake and preference.
Collapse
Affiliation(s)
- Bailey N. Keller
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Angela E. Snyder
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Caitlin R. Coker
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Elizabeth A. Aguilar
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Mary K. O’Brien
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Sarah S. Bingaman
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Amy C. Arnold
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Andras Hajnal
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| | - Yuval Silberman
- The Pennsylvania State University College of Medicine, Department of Neural and Behavioral Sciences
| |
Collapse
|
41
|
Agoitia A, Cruz-Sanchez A, Balderas I, Bermúdez-Rattoni F. The anterior insula and its projection to amygdala nuclei modulate the abstinence-exacerbated expression of conditioned place preference. Psychopharmacology (Berl) 2024; 241:445-459. [PMID: 38010515 PMCID: PMC10884150 DOI: 10.1007/s00213-023-06499-0] [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: 09/13/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
RATIONALE Relapse into substance use is often triggered by exposure to drug-related environmental cues. The magnitude of drug seeking depends on the duration of abstinence, a phenomenon known as the incubation of drug craving. Clinical and preclinical research shows that the insular cortex is involved in substance use disorders and cue-induced drug seeking. However, the role of the insula on memory retrieval and motivational integration for cue-elicited drug seeking remains to be determined. OBJECTIVES We investigated the role of the anterior insular cortex (aIC) and its glutamatergic projection to amygdala nuclei (aIC-AMY) on the expression of conditioned place preference (CPP) during early and late abstinence. METHODS Male adult C57BL/6J mice underwent amphetamine-induced CPP, and their preference was tested following 1 or 14 days of abstinence. aIC and aIC-AMY functional role in CPP expression was assessed at both abstinence periods by employing optogenetic silencing and behavioral pharmacology. RESULTS Compared to a single day, an exacerbated preference for the amphetamine-paired context was observed after 14 days of abstinence. Photoinhibition of either aIC or aIC-AMY projection reduced CPP expression following late but not early abstinence. Similarly, the antagonism of aIC NMDA receptors reduced CPP expression after 14 days of abstinence but not 1 day. CONCLUSIONS These results suggest that aIC and its glutamatergic output to amygdala nuclei constitute critical neurobiological substrates mediating enhanced motivational cue reactivity during the incubation of amphetamine craving rather than contextual memory recall. Moreover, cortical NMDA receptor signaling may become sensitized during abstinence, ultimately modulating disproportioned drug seeking.
Collapse
Affiliation(s)
- Andrés Agoitia
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Apolinar Cruz-Sanchez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Israela Balderas
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Federico Bermúdez-Rattoni
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
42
|
Lee JH, Kang S, Maier SU, Lee SA, Goldfarb EV, Ahn WY. Acute Stress Enhances Memory and Preference for Smoking-Related Associations in Smokers. Nicotine Tob Res 2024; 26:333-341. [PMID: 37589502 DOI: 10.1093/ntr/ntad152] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Nicotine dependence follows a chronic course that is characterized by repeated relapse, often driven by acute stress and rewarding memories of smoking retrieved from related contexts. These two triggers can also interact, with stress influencing retrieval of contextual memories. However, the roles of these processes in nicotine dependence remain unknown. AIMS AND METHODS We investigated how acute stress biases memory for smoking-associated contexts among smokers (N = 65) using a novel laboratory paradigm. On day 1, participants formed associations between visual stimuli of items (either neutral or related to smoking) and places (background scenes). On day 2 (24 hours later), participants were exposed to an acute laboratory-based stressor (socially evaluated cold pressor test; N = 32) or a matched control condition (N = 33) prior to being tested on their memory recognition and preferences for each item and place. We distinguished the accuracy of memory into specific (ie, precisely correct) or gist (ie, lure items with similar content) categories. RESULTS Results demonstrated that the stressor significantly induced physiological and subjective perceived stress responses, and that stressed smokers exhibited a memory bias in favor of smoking-related items. In addition, the stressed group displayed greater preference for both smoking-related items and places that had been paired with the smoking-related items. We also found suggestive evidence that stronger smoking-related memory biases were associated with more severe nicotine dependence (ie, years of smoking). CONCLUSIONS These results highlight the role of stress in biasing smokers toward remembering contexts associated with smoking, and amplifying their preference for these contexts. IMPLICATIONS The current study elucidates the role of acute stress in promoting memory biases favoring smoking-related associations among smokers. The results suggest that the retrieval of smoking-biased associative memory could be a crucial factor in stress-related nicotine seeking. This may lead to a potential intervention targeting the extinction of smoking-related context memories as a preventive strategy for stress-induced relapse.
Collapse
Affiliation(s)
- Jeung-Hyun Lee
- Department of Psychology, Seoul National University, Seoul, South Korea
| | - Sanghoon Kang
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA
| | - Silvia U Maier
- Translational Neuromodeling Unit, University of Zurich, Zürich, Switzerland
| | - Sang Ah Lee
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| | | | - Woo-Young Ahn
- Department of Psychology, Seoul National University, Seoul, South Korea
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea
| |
Collapse
|
43
|
Hleihil M, Benke D. Restoring GABA B receptor expression in the ventral tegmental area of methamphetamine addicted mice inhibits locomotor sensitization and drug seeking behavior. Front Mol Neurosci 2024; 17:1347228. [PMID: 38384279 PMCID: PMC10879384 DOI: 10.3389/fnmol.2024.1347228] [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: 11/30/2023] [Accepted: 01/15/2024] [Indexed: 02/23/2024] Open
Abstract
Repeated exposure to psychostimulants such as methamphetamine (METH) induces neuronal adaptations in the mesocorticolimbic dopamine system, including the ventral tegmental area (VTA). These changes lead to persistently enhanced neuronal activity causing increased dopamine release and addictive phenotypes. A factor contributing to increased dopaminergic activity in this system appears to be reduced GABAB receptor-mediated neuronal inhibition in the VTA. Dephosphorylation of serine 783 (Ser783) of the GABAB2 subunit by protein phosphatase 2A (PP2A) appears to trigger the downregulation GABAB receptors in psychostimulant-addicted rodents. Therefore, preventing the interaction of GABAB receptors with PP2A using an interfering peptide is a promising strategy to restore GABAB receptor-mediated neuronal inhibition. We have previously developed an interfering peptide (PP2A-Pep) that inhibits the GABAB receptors/PP2A interaction and thereby restores receptor expression under pathological conditions. Here, we tested the hypothesis that restoration of GABAB receptor expression in the VTA of METH addicted mice reduce addictive phenotypes. We found that the expression of GABAB receptors was significantly reduced in the VTA and nucleus accumbens but not in the hippocampus and somatosensory cortex of METH-addicted mice. Infusion of PP2A-Pep into the VTA of METH-addicted mice restored GABAB receptor expression in the VTA and inhibited METH-induced locomotor sensitization as assessed in the open field test. Moreover, administration of PP2A-Pep into the VTA also reduced drug seeking behavior in the conditioned place preference test. These observations underscore the importance of VTA GABAB receptors in controlling addictive phenotypes. Furthermore, this study illustrates the value of interfering peptides targeting diseases-related protein-protein interactions as an alternative approach for a potential development of selective therapeutic interventions.
Collapse
Affiliation(s)
- Mohammad Hleihil
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
| | - Dietmar Benke
- Institute of Pharmacology and Toxicology, University of Zurich, Zürich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zürich, Switzerland
| |
Collapse
|
44
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [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] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
45
|
McDevitt DS, Wade QW, McKendrick GE, Nelsen J, Starostina M, Tran N, Blendy JA, Graziane NM. The Paraventricular Thalamic Nucleus and Its Projections in Regulating Reward and Context Associations. eNeuro 2024; 11:ENEURO.0524-23.2024. [PMID: 38351131 PMCID: PMC10883411 DOI: 10.1523/eneuro.0524-23.2024] [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: 12/11/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
The paraventricular thalamic nucleus (PVT) is a brain region that mediates aversive and reward-related behaviors as shown in animals exposed to fear conditioning, natural rewards, or drugs of abuse. However, it is unknown whether manipulations of the PVT, in the absence of external factors or stimuli (e.g., fear, natural rewards, or drugs of abuse), are sufficient to drive reward-related behaviors. Additionally, it is unknown whether drugs of abuse administered directly into the PVT are sufficient to drive reward-related behaviors. Here, using behavioral as well as pathway and cell-type specific approaches, we manipulate PVT activity as well as the PVT-to-nucleus accumbens shell (NAcSh) neurocircuit to explore reward phenotypes. First, we show that bath perfusion of morphine (10 µM) caused hyperpolarization of the resting membrane potential, increased rheobase, and decreased intrinsic membrane excitability in PVT neurons that project to the NAcSh. Additionally, we found that direct injections of morphine (50 ng) in the PVT of mice were sufficient to generate conditioned place preference (CPP) for the morphine-paired chamber. Mimicking the inhibitory effect of morphine, we employed a chemogenetic approach to inhibit PVT neurons that projected to the NAcSh and found that pairing the inhibition of these PVT neurons with a specific context evoked the acquisition of CPP. Lastly, using brain slice electrophysiology, we found that bath-perfused morphine (10 µM) significantly reduced PVT excitatory synaptic transmission on both dopamine D1 and D2 receptor-expressing medium spiny neurons in the NAcSh, but that inhibiting PVT afferents in the NAcSh was not sufficient to evoke CPP.
Collapse
Affiliation(s)
- Dillon S McDevitt
- Neuroscience Program, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Quinn W Wade
- Department of Anesthesiology and Perioperative Medicine, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Greer E McKendrick
- Neuroscience Program, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Jacob Nelsen
- Doctor of Medicine Program, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Mariya Starostina
- Doctor of Medicine Program, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Nam Tran
- Doctor of Medicine Program, Penn State College of Medicine, Hershey, Pennsylvania 17033
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Nicholas M Graziane
- Departments of Anesthesiology and Perioperative Medicine and Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania 17033
| |
Collapse
|
46
|
Abboussi O, Khan ZA, Ibork H, Zulu SS, Daniels W, Taghzouti K, Hales TG. CB2 agonist mitigates cocaine-induced reinstatement of place preference and modulates the inflammatory response in mice. Behav Pharmacol 2024; 35:26-35. [PMID: 38085651 DOI: 10.1097/fbp.0000000000000759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Chronic exposure to cocaine is known to have profound effects on the brain, leading to the dysregulation of inflammatory signalling pathways, the activation of microglia, and the manifestation of cognitive and motivational behavioural impairments. The endocannabinoid system has emerged as a potential mediator of cocaine's deleterious effects. In this study, we sought to investigate the therapeutic potential of the cannabinoid CB2 receptor agonist, JWH-133, in mitigating cocaine-induced inflammation and associated motivational behavioural alterations in an in vivo model. Our research uncovered compelling evidence that JWH-133, a selective CB2 receptor agonist, exerts a significant dampening effect on the reinstatement of cocaine-induced conditioned place preference. This effect was accompanied by notable changes in the neurobiological landscape. Specifically, JWH-133 administration was found to upregulate Δ-FOSB expression in the nucleus accumbens (Nac), elevate CX3CL1 levels in both the ventral tegmental area and prefrontal cortex (PFC), and concurrently reduce IL-1β expression in the PFC and NAc among cocaine-treated animals. These findings highlight the modulatory role of CB2 cannabinoid receptor activation in altering the reward-seeking behaviour induced by cocaine. Moreover, they shed light on the intricate interplay between the endocannabinoid system and cocaine-induced neurobiological changes, paving the way for potential therapeutic interventions targeting CB2 receptors in the context of cocaine addiction and associated behavioural deficits.
Collapse
Affiliation(s)
- Oualid Abboussi
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Zmarak Ahmad Khan
- Institute of Academic Anaesthesia, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hind Ibork
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Simo S Zulu
- Department of Human Biology, Faculty of Health Sciences, Nelson Mandela University, Port Elisabeth
| | - William Daniels
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Khalid Taghzouti
- Physiology and Physiopathology Team, Faculty of Sciences, Genomic of Human Pathologies Research Centre, Mohammed V University in Rabat, Rabat, Morocco
| | - Tim G Hales
- Institute of Academic Anaesthesia, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
47
|
Alvarado-Torres JK, Morales-Silva R, Sanabria Ponce de Leon A, Rodriguez-Torres G, Perez-Torres J, Perez-Perez Y, Mueller D, Sepulveda-Orengo MT. Estradiol reduction through aromatase inhibition impairs cocaine seeking in male rats. Front Behav Neurosci 2024; 17:1307606. [PMID: 38292056 PMCID: PMC10824998 DOI: 10.3389/fnbeh.2023.1307606] [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: 10/04/2023] [Accepted: 12/26/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction Clinical and preclinical research on cocaine use disorder (CUD) has shown that sex differences in drug seeking are influenced by hormonal fluctuations. Estradiol (E2), a sex steroid hormone, has been linked to female drug effects, vulnerability to use/abuse, and psychosocial factors. Preclinical studies show that estradiol in females facilitates the extinction of cocaine-seeking behavior indicating a possible role in regulating extinction learning. Similar to females, males' brains contain the aromatase enzyme which converts testosterone to estradiol. However, it is unclear whether estradiol plays a role in male extinction learning as it does in females. Furthermore, how endogenously aromatized estradiol affects drug addiction in males is unknown. Therefore, this study investigated whether endogenous estradiol regulates cocaine seeking in male rats. We hypothesized that decreased aromatase enzyme activity, resulting in decreased estradiol synthesis in male brains, will impair extinction learning leading to increased cocaine-seeking behavior. Methods This hypothesis was tested using cocaine-conditioned place preference (CPP), and short access self-administration (SA), followed by extinction and reinstatement. Before each extinction session for CPP or SA, male rats received an injection of either 1 (low dose) or 2.5 mg/kg (high dose) of the aromatase inhibitor Fadrozole (FAD), or vehicle. Results FAD groups showed dose-dependent effects on cocaine-seeking behavior compared to the vehicle group during CPP extinction. Specifically, low dose FAD facilitated extinction of cocaine CPP, whereas high dose FAD impaired it. In contrast, neither dose of FAD had any effects on the extinction of cocaine SA. Interestingly, only the low dose FAD group had decreased active lever pressing during cue- and cocaine-primed reinstatement compared to the vehicle group. Neither dose of FAD had an effect on sucrose extinction or reinstatement of sucrose seeking. Discussion These results from CPP experiments suggest that estradiol may impact extinction learning, as a low dose of FAD may strengthen the formation of cocaine extinction memory. Additionally, in male rats undergoing cocaine SA, the same low dose of aromatase inhibitor effectively reduced reinstatement of cocaine-seeking behavior. Thus, estradiol impacts cocaine seeking and extinction in both males and females, and it may also influence the development of sex-specific treatment strategies for CUD.
Collapse
Affiliation(s)
- John K. Alvarado-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Roberto Morales-Silva
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | | | - Genesis Rodriguez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Joshua Perez-Torres
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Yobet Perez-Perez
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Devin Mueller
- Department of Biological Sciences, Kent State University, Kent, OH, United States
| | - Marian T. Sepulveda-Orengo
- Basic Sciences Department, Ponce Research Institute, Ponce Health Sciences University, Ponce, Puerto Rico
| |
Collapse
|
48
|
Yalniz Y, Yunusoğlu O, Berköz M, Demirel ME. Effects of fisetin on ethanol-induced rewarding properties in mice. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2024; 50:75-83. [PMID: 38235981 DOI: 10.1080/00952990.2023.2292976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/05/2023] [Indexed: 01/19/2024]
Abstract
Background: Alcohol use disorder (AUD) is a chronic relapsing disorder associated with compulsive drinking of alcohol. Natural flavonoid fisetin affects a variety of transmitter systems relevant to AUD, such as aminobutyric acid, N-methyl-D-aspartate, and dopamine, as well as peroxisome proliferator-activated receptors.Objectives: This study investigated fisetin's impact on the motivational properties of ethanol using conditioned place preference (CPP) in mice (n = 50).Methods: Mice were conditioned with ethanol (2 g/kg, i.p.) or saline on alternating days for 8 consecutive days and were given intragastric (i.g.) fisetin (10, 20, or 30 mg/kg, i.g.), 45 min before ethanol conditioning. During extinction, physiological saline was injected to the control and ethanol groups, and fisetin was administered to the fisetin groups. To evaluate the effect of fisetin on the reinstatement of ethanol-induced CPP, fisetin was given 45 min before a priming dose of ethanol (0.4 g/kg, i.p.; reinstatement test day).Results: Fisetin decreased the acquisition of ethanol-induced CPP (30 mg/kg, p < .05) and accelerated extinction (20 and 30 mg/kg, p < .05). Furthermore, fisetin attenuated reinstatement of ethanol-induced CPP (30 mg/kg, p < .05).Conclusions: Fisetin appears to diminish the rewarding properties of ethanol, as indicated by its inhibitory effect and facilitation of extinction in ethanol-induced CPP. These findings imply a potential therapeutic application of fisetin in preventing ethanol-seeking behavior, promoting extinction, and reducing the risk of relapse.
Collapse
Affiliation(s)
- Yasin Yalniz
- Department of Pharmacology, Faculty of Medicine, Bolu Izzet Baysal University, Bolu, Turkey
| | - Oruç Yunusoğlu
- Department of Pharmacology, Faculty of Medicine, Bolu Izzet Baysal University, Bolu, Turkey
| | - Mehmet Berköz
- Department of Biochemistry, Faculty of Pharmacy, Van Yuzuncu Yıl University, Van, Turkey
| | - Mustafa Enes Demirel
- Emergency Department, School of Medicine, Bolu Abant Izzet Baysal University, Bolu, Turkey
| |
Collapse
|
49
|
Saito A, Murata H, Niitani K, Nagasaki J, Otoda A, Chujo Y, Yanagida J, Nishitani N, Deyama S, Kaneda K. Social defeat stress enhances the rewarding effects of cocaine through α 1A adrenoceptors in the medial prefrontal cortex of mice. Neuropharmacology 2024; 242:109757. [PMID: 37839511 DOI: 10.1016/j.neuropharm.2023.109757] [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: 08/05/2023] [Revised: 10/07/2023] [Accepted: 10/07/2023] [Indexed: 10/17/2023]
Abstract
Various stressors potentiate the rewarding effects of cocaine and contribute to cocaine cravings. However, it remains unclear whether psychosocial stress enhances the rewarding effects of cocaine. Accordingly, this study employed a cocaine-conditioned place preference (CPP) paradigm combined with social defeat (SD) exposure to investigate the effects of acute SD stress on cocaine reward in male mice. We found that SD stress immediately before the posttest significantly increased cocaine CPP, and systemic blockade of α1 adrenoceptors, but not β adrenoceptors, suppressed this increase. Fiber photometry recordings with GRABNE1m sensors revealed increased noradrenaline (NA) levels in the medial prefrontal cortex (mPFC) in test mice in response to attacks by aggressor mice during SD. Moreover, the SD stress-induced enhancement of CPP was effectively suppressed by intra-mPFC infusion of an α1 adrenoceptor antagonist. In vitro whole-cell recordings demonstrated that silodosin, an α1A, but not α1B or α1D, adrenoceptor antagonist, inhibited NA-induced depolarizing currents and facilitation of excitatory synaptic transmissions. Consistently, intra-mPFC silodosin infusion significantly suppressed the SD stress-induced CPP enhancement. Conversely, intra-mPFC infusion of α1A adrenoceptor agonist augmented cocaine CPP in the absence of stress exposure. Additionally, intranasal silodosin administration attenuated the SD stress-induced enhancement of CPP, and chemogenetic inhibition of mPFC excitatory neurons also suppressed the SD stress-induced CPP enhancement. Together, these findings suggest that NA stimulation of α1A adrenoceptors and the subsequent activation of mPFC pyramidal cells may contribute to SD stress-induced amplification of the rewarding effects of cocaine, and intranasal silodosin administration may hold therapeutic potential for mitigating stress-associated cocaine craving.
Collapse
Affiliation(s)
- Atsushi Saito
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Haruka Murata
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Kazuhei Niitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Junpei Nagasaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Atsuki Otoda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusuke Chujo
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Junko Yanagida
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
50
|
Aguilar LA, Coker CR, McCullers Z, Evans A, Showemimo O, Melkumyan M, Keller BN, Snyder AE, Bingaman SS, Randall PA, Hajnal A, Browning KN, Arnold AC, Silberman Y. Adolescent alcohol disrupts development of noradrenergic neurons in the nucleus of the tractus solitarius and enhances stress behaviors in adulthood in mice in a sex specific manner. ADDICTION NEUROSCIENCE 2023; 9:100132. [PMID: 38162404 PMCID: PMC10756564 DOI: 10.1016/j.addicn.2023.100132] [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: 01/03/2024]
Abstract
Alcohol use disorders (AUDs) are common mental health issues worldwide and can lead to other chronic diseases. Stress is a major factor in the development and continuation of AUDs, and adolescent alcohol exposure can lead to enhanced stress-responsivity and increased risk for AUD development in adulthood. The exact mechanisms behind the interaction between adolescence, stress, and alcohol are not fully understood and require further research. In this regard, the nucleus of the tractus solitarius (NTS) provides dense norepinephrine projections to the extended amygdala, providing a key pathway for stress-related alcohol behaviors. While NTS norepinephrine neurons are known to be alcohol sensitive, whether adolescent alcohol disrupts NTS-norepinephrine neuron development and if this is related to altered stress-sensitivity and alcohol preference in adulthood has not previously been examined. Here, we exposed male and female C57Bl/6J mice to the commonly used adolescent intermittent ethanol (AIE) vapor model during postnatal day 28-42 and examined AIE effects on: 1) tyrosine hydroxylase (TH) mRNA expression in the NTS across various ages (postnatal day 21-90), 2) behavioral responses to acute stress in the light/dark box test in adulthood, 3) NTS TH neuron responses to acute stress and ethanol challenges in adulthood, and 4) ethanol conditioned place preference behavior in adulthood. Overall the findings indicate that AIE alters NTS TH mRNA expression and increases anxiety-like behaviors following acute stress exposure in a sex-dependent manner. These mRNA expression and behavioral changes occur in the absence of AIE-induced changes in NTS TH neuron sensitivity to either acute stress or acute alcohol exposure or changes to ethanol conditioned place preference.
Collapse
Affiliation(s)
- Liz A. Aguilar
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Currently at Department of Biology, Indiana University Bloomington, USA
| | - Caitlin R. Coker
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Anatomy, USA
- Currently at Georgetown University School of Medicine, USA
| | - Zari McCullers
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Biomedical Sciences, USA
| | - Alexandra Evans
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Biomedical Sciences, USA
| | - Opeyemi Showemimo
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Anatomy, USA
| | - Mariam Melkumyan
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Bailey N. Keller
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Angela E. Snyder
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
- Penn State College of Medicine, Graduate Program in Neuroscience, USA
| | - Sarah S. Bingaman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | | | - Andras Hajnal
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Amy C. Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| | - Yuval Silberman
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, USA
| |
Collapse
|