1
|
Miguel Telega L, Ashouri Vajari D, Ramanathan C, Coenen VA, Döbrössy MD. Chronic in vivo sequelae of repetitive acute mfb-DBS on accumbal dopamine and midbrain neuronal activity. J Neurochem 2025; 169:e16223. [PMID: 39308085 DOI: 10.1111/jnc.16223] [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: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 12/20/2024]
Abstract
Medial Forebrain Bundle Deep Brain Stimulation (MFB-DBS) can have rapid and long lasting antidepressant effects in Treatment Resistant Depression (TRD) patients. The mechanisms are not well understood, but one hypothesis stipulates that modulation of the dopaminergic (DAergic) fibers contribute to the therapeutic outcome. Acute DBS effects on DA release have been studied; however, longitudinal studies with acute-repetitive DBS are lacking. Long-Evans accumbal DA release and Ventral Tegmental Area (VTA) calcium tonic and phasic signaling to different mfb-DBS parameters were measured using fiber photometry over 8 weeks, following acute and repetitive stimulation in behaving and non-behaving animals. DBS-induced release was observed in both targets, with increased frequency and DBS duration. 130 Hz stimulation increased phasic and tonic DA response over time, with the latter being a potential mechanism for its long-term clinical effectiveness. VTA calcium transients decreased, while phasic activity increased with frequency. Pulse width (PW)-mediated differential peak release timing also suggests potential parallel activation of diverse fiber types. Additionally, decreased DA transients rate during Elevated Plus Maze (EPM) suggests context and stimulation duration-dependent DA release. The data confirm chronic antidromic/orthodromic DAergic responses with stimulation parameter dependent variability, providing novel insights into temporal adaptations, connectivity and fiber recruitment on mfb DBS.
Collapse
Affiliation(s)
- Lidia Miguel Telega
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
| | - Danesh Ashouri Vajari
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
- Laboratory for Biomedical Microtechnology, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg im Breisgau, Germany
| | - Chockalingam Ramanathan
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- Institute for Physiology I, Medical Faculty, Albert-Ludwigs-University Freiburg, Freiburg im Breisgau, Germany
- Bernstein Center Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- BrainLinks-BrainTools, IMBIT (Institute for Machine-Brain Interfacing Technology), University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
- Center for Basics in Neuromodulation, University of Freiburg, Freiburg im Breisgau, Germany
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences (SIN), Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center, -University of Freiburg, Freiburg im Breisgau, Germany
- Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
- Center for Basics in Neuromodulation, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
2
|
Yang J, Li X, Gao Y, Wang W, Wang J, Wang Y, Gong C, Wang R, Yuan L. The influential factors of depression among caregivers of children with Cleft Lip and/or Palate: a path analysis based on diathesis-stress theoretical model. BMC Psychiatry 2024; 24:911. [PMID: 39696081 DOI: 10.1186/s12888-024-06383-x] [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: 07/03/2023] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Depression is the most common psychological phenomenon among caregivers of children with Cleft Lip and/or Palate and affects the quality of life, treatment satisfaction, children's self-esteem and parent-child relationship. However, Existing studies have limited explanations for the mechanisms that lead to depression in caregivers of children with Cleft Lip and/or Palate. The present study aimed to identify factors influencing depression among caregivers of children with Cleft Lip and/or Palate and to provide a reference point and new ideas for future research. METHODS This cross-sectional study investigated 248 caregivers at two Cleft Lip and/or Palate treatment centers in China, spanning from April 2019 to July 2020. The diathesis-stress model was used to construct the model, and data analysis was conducted using SPSS 26.0 and AMOS 25.0. Path analysis was employed to test the hypothetical model, and the fit of the model was evaluated using Chi-Square/degree of freedom, Goodness-of-Fit Index, Normed Fit Index, Relative Fit Index, Incremental Fit Index, Tacker-Lewis Index, Comparative Fit Index and Root Mean Square Error of Approximation. RESULTS The study found that the average level of depression among caregivers of children with Cleft Lip and/or Palate was 4.83 (4.79). Additionally, 43.5% of caregivers exhibited depressive symptoms. The fit of the modified path model was found to be satisfactory, with the following indices: Chi-Square/degree of freedom = 1.881, Goodness-of-Fit Index = 0.986, Normed Fit Index = 0.973, Relative Fit Index = 0.919, Incremental Fit Index = 0.987, Tucker-Lewis Index = 0.960, Comparative Fit Index = 0.987, and Root Mean Square Error of Approximation (RMSEA) = 0.060. The results indicated that parenting stress had the greatest direct impact on depression, followed by hope. Indirect effects on depression were observed for optimism, resilience, and coping, which were mediated through perceived social support and parenting stress. CONCLUSIONS This study suggests that depression in caregivers of children with Cleft Lip and/or Palate was leadingly influenced by parenting stress and hope. Strategies aimed at the key factors are expected to decrease depression.
Collapse
Affiliation(s)
- Jinrong Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaohan Li
- School of Nursing, China Medical University, Shenyang, China
| | - Yuqin Gao
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Weiren Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Junyan Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Yanjie Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Caixia Gong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Runzi Wang
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China.
| | - Lulu Yuan
- School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China.
| |
Collapse
|
3
|
Parker KE, Kuo CC, Buckley AR, Patterson AP, Duong V, Hunter SC, McCall JG. Monosynaptic ventral tegmental area glutamate projections to the locus coeruleus enhance aversive processing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.615025. [PMID: 39713345 PMCID: PMC11661122 DOI: 10.1101/2024.10.04.615025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Distinct excitatory synaptic inputs to the locus coeruleus (LC) modulate behavioral flexibility. Here we identify a novel monosynaptic glutamatergic input to the LC from the ventral tegmental area (VTA). We show robust VTA axonal projections provide direct glutamatergic transmission to LC. Despite weak synaptic summation, optogenetic activation of these axons enhances LC tonic firing and facilitates real-time and conditioned aversive behaviors. We hypothesized this projection may modulate synaptic integration with other excitatory inputs. We then used coincident VTA-LC photostimulation with local electrical stimulation and observed enhanced LC burst induction. To determine whether this integration also occurs in vivo, we took an analogous approach measuring reward-seeking behavior during unpredictable probabilistic punishment. Here, glutamatergic VTA-LC photostimulation during a concurrent noxious stimulus did not delay reward-seeking behavior, but increased probability of task failure. Together, we identified a novel VTA-LC glutamatergic projection that drives concurrent synaptic summation during salient stimuli to promote behavioral avoidance.
Collapse
Affiliation(s)
- Kyle E. Parker
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Chao-Cheng Kuo
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Alex R. Buckley
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Abigail P. Patterson
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Vincent Duong
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarah C. Hunter
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jordan G. McCall
- Department of Anesthesiology, Center for Clinical Pharmacology, Washington University Pain Center, Washington University School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
4
|
Jiang Q, Bakhurin KI, Hughes RN, Lu B, Ruan S, Yin HH. GABAergic neurons from the ventral tegmental area represent and regulate force vectors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627361. [PMID: 39713374 PMCID: PMC11661075 DOI: 10.1101/2024.12.07.627361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
The ventral tegmental area (VTA), a midbrain region associated with motivated behaviors, consists predominantly of dopaminergic (DA) neurons and GABAergic (GABA) neurons. Previous work has suggested that VTA GABA neurons provide a reward prediction, which is used in computing a reward prediction error. In this study, using in vivo electrophysiology and continuous quantification of force exertion in head-fixed mice, we discovered distinct populations of VTA GABA neurons that exhibited precise force tuning independently of learning, reward prediction, and outcome valence. Their activity usually preceded force exertion, and selective optogenetic manipulations of these neurons systematically modulated force exertion without influencing reward prediction. Together, these findings show that VTA GABA neurons continuously regulate force vectors during motivated behavior.
Collapse
|
5
|
Stelzner ME, Wolff AR, Saunders BT. Ventral tegmental area GABA neurons integrate positive and negative valence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.07.627330. [PMID: 39677805 PMCID: PMC11642998 DOI: 10.1101/2024.12.07.627330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Ventral tegmental area (VTA) dopamine (DA) neurons are classically linked to Pavlovian reward learning and reinforcement. Intermingled VTA GABA neurons are positioned to regulate dopaminergic and striatal systems, but we lack critical insight into how this population contributes to conditioned motivation in different learning contexts. Recording DA and GABA neurons across multiple conditioning paradigms, we found that GABA neurons not only actively encode appetitive and aversive cues and outcomes separately, but uniquely integrate salient events of both valences to guide reward seeking.
Collapse
Affiliation(s)
- Margaret E Stelzner
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
- Graduate Program in Neuroscience, University of Minnesota
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| |
Collapse
|
6
|
Jia W, Peng J, Zhang Y, Zhu J, Qiang X, Zhang R, Shi L. Amelioration impact of gut-brain communication on obesity control by regulating gut microbiota composition through the ingestion of animal-plant-derived peptides and dietary fiber: can food reward effect as a hidden regulator? Crit Rev Food Sci Nutr 2024; 64:11575-11589. [PMID: 37526310 DOI: 10.1080/10408398.2023.2241078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Various roles of intestinal flora in the gut-brain axis response pathway have received enormous attention because of their unique position in intestinal flora-derived metabolites regulating hormones, inducing appetite, and modulating energy metabolism. Reward pathways in the brain play a crucial role in gut-brain communications, but the mechanisms have not been methodically understood. This review outlined the mechanisms by which leptin, ghrelin, and insulin are influenced by intestinal flora-derived metabolites to regulate appetite and body weight, focused on the significance of the paraventricular nucleus and ventromedial prefrontal cortex in food reward. The vagus nerve and mitochondria are essential pathways of the intestinal flora involved in the modulation of neurotransmitters, neural signaling, and neurotransmission in gut-brain communications. The dynamic response to nutrient intake and changes in the characteristics of feeding activity requires the participation of the vagus nerve to transmit messages to be completed. SCFAs, Bas, BCAAs, and induced hormones mediate the sensory information and reward signaling of the host in the complex regulatory mechanism of food selection, and the composition of the intestinal flora significantly impacts this process. Food reward in the process of obesity based on gut-brain communications expands new ideas for the prevention and treatment of obesity.
Collapse
Affiliation(s)
- Wei Jia
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
- Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an, China
- Shaanxi Sky Pet Biotechnology Co., Ltd, Xi'an, China
| | - Jian Peng
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yan Zhang
- Inspection and Testing Center of Fuping County (Shaanxi Goat Milk Product Quality Supervision and Inspection Center), Wei nan, China
| | - Jiying Zhu
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Xin Qiang
- Inspection and Testing Center of Fuping County (Shaanxi Goat Milk Product Quality Supervision and Inspection Center), Wei nan, China
| | - Rong Zhang
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Lin Shi
- School of Food and Bioengineering, Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
7
|
Lefner MJ, Moghaddam B. Reward and punishment contingency shifting reveals distinct roles for VTA dopamine and GABA neurons in behavioral flexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.07.617060. [PMID: 39416212 PMCID: PMC11482802 DOI: 10.1101/2024.10.07.617060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
In dynamic environments where stimuli predicting rewarding or aversive outcomes unexpectedly change, it is critical to flexibly update behavior while preserving recollection of previous associations. Dopamine and GABA neurons in the ventral tegmental area (VTA) are implicated in reward and punishment learning, yet little is known about how each population adapts when the predicted outcome valence changes. We measured VTA dopamine and GABA population activity while male and female rats learned to associate three discrete auditory cues to three distinct outcomes: reward, punishment, or no outcome within the same session. After learning, the reward and punishment cue-outcome contingencies were reversed, and subsequently re-reversed. As expected, the dopamine population rapidly adapted to learning and contingency reversals by increasing the response to appetitive stimuli and decreasing the response to aversive stimuli. In contrast, the GABA population increased activity to all sensory events regardless of valence, including the neutral cue. Reversing learned contingencies selectively influenced GABA responses to the reward-predictive cue, prolonging increased activity within and across sessions. The observed valence-specific dissociations in the directionality and temporal progression of VTA dopamine and GABA calcium activity indicates that these populations are independently recruited and serve distinct roles during appetitive and aversive associative learning and contingency reversal.
Collapse
Affiliation(s)
- Merridee J Lefner
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR
| | - Bita Moghaddam
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR
| |
Collapse
|
8
|
Ebrahimi MN, Banazadeh M, Alitaneh Z, Jaafari Suha A, Esmaeili A, Hasannejad-Asl B, Siahposht-Khachaki A, Hassanshahi A, Bagheri-Mohammadi S. The distribution of neurotransmitters in the brain circuitry: Mesolimbic pathway and addiction. Physiol Behav 2024; 284:114639. [PMID: 39004195 DOI: 10.1016/j.physbeh.2024.114639] [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: 04/23/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
Understanding the central nervous system (CNS) circuitry and its different neurotransmitters that underlie reward is essential to improve treatment for many common health issues, such as addiction. Here, we concentrate on understanding how the mesolimbic circuitry and neurotransmitters are organized and function, and how drug exposure affects synaptic and structural changes in this circuitry. While the role of some reward circuits, like the cerebral dopamine (DA)/glutamate (Glu)/gamma aminobutyric acid (GABA)ergic pathways, in drug reward, is well known, new research using molecular-based methods has shown functional alterations throughout the reward circuitry that contribute to various aspects of addiction, including craving and relapse. A new understanding of the fundamental connections between brain regions as well as the molecular alterations within these particular microcircuits, such as neurotrophic factor and molecular signaling or distinct receptor function, that underlie synaptic and structural plasticity evoked by drugs of abuse has been made possible by the ability to observe and manipulate neuronal activity within specific cell types and circuits. It is exciting that these discoveries from preclinical animal research are now being applied in the clinic, where therapies for human drug dependence, such as deep brain stimulation and transcranial magnetic stimulation, are being tested. Therefore, this chapter seeks to summarize the current understanding of the important brain regions (especially, mesolimbic circuitry) and neurotransmitters implicated in drug-related behaviors and the molecular mechanisms that contribute to altered connectivity between these areas, with the postulation that increased knowledge of the plasticity within the drug reward circuit will lead to new and improved treatments for addiction.
Collapse
Affiliation(s)
- Mohammad Navid Ebrahimi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Banazadeh
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Zahra Alitaneh
- Quantitative and System Biology, Department of Natural Sciences, University of California Merced, USA
| | - Ali Jaafari Suha
- Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Esmaeili
- Student Research Committee, Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Hasannejad-Asl
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti, University of Medical Sciences, Tehran, Iran
| | - Ali Siahposht-Khachaki
- Immunogenetics Research Center, Department of Physiology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amin Hassanshahi
- Department of Physiology, Bam University of Medical Sciences, Bam, Iran
| | - Saeid Bagheri-Mohammadi
- Department of Paramedicine, Amol School of Paramedical Sciences, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
9
|
Pearson AC, Ostroumov A. Midbrain KCC2 downregulation: Implications for stress-related and substance use behaviors. Curr Opin Neurobiol 2024; 88:102901. [PMID: 39142020 PMCID: PMC11392611 DOI: 10.1016/j.conb.2024.102901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
Stress-related and substance use disorders are both characterized by disruptions in reward-related behaviors, and these disorders are often comorbid with one another. Recent investigations have identified a novel mechanism of inhibitory plasticity induced by both stress and substance use within the ventral tegmental area (VTA), a key region in reward processing. This mechanism involves the neuron-specific potassium chloride cotransporter isoform 2 (KCC2), which is essential in modulating inhibitory signaling through the regulation of intracellular chloride (Cl-) in VTA GABA neurons. Experiences, such as exposure to stress or substance use, diminish KCC2 expression in VTA GABA neurons, leading to abnormal reward-related behaviors. Here, we review literature suggesting that KCC2 downregulation contributes to irregular dopamine (DA) transmission, impacting multiple reward circuits and promoting maladaptive behaviors. Activating KCC2 restores canonical GABA functioning and reduces behavioral deficits in preclinical models, leading us to advocate for KCC2 as a target for therapies aimed at alleviating and mitigating various stress-related and substance use disorders.
Collapse
Affiliation(s)
- Anna C Pearson
- Department of Pharmacology & Physiology, Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20057, USA. https://twitter.com/AnnaCPearson
| | - Alexey Ostroumov
- Department of Pharmacology & Physiology, Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20057, USA.
| |
Collapse
|
10
|
Feng AY, Barbosa DAN, Casey AB, Rijsketic DR, Salgado JS, Huang H, Malenka RC, Hermes D, Miller KJ, Halpern CH, Heifets BD. Cross-species brain-wide mapping reveals a conserved and coordinated network engaged by NAc DBS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.08.611940. [PMID: 39314466 PMCID: PMC11419029 DOI: 10.1101/2024.09.08.611940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Nucleus accumbens (NAc) deep brain stimulation (DBS) has been increasingly explored as a treatment modality for refractory neuropsychiatric disorders. Uncovering the accumbens network that is engaged by DBS is a critical step forward in understanding how modulating this important node impacts the broader mesocorticolimbic circuit. Using whole-brain clearing and unbiased, brain-wide neural activity mapping, we found that NAc DBS increases neural activity in a coordinated mesocorticolimbic network in mice. Simultaneous intracranial electrophysiology recordings from the human NAc and brief stimulation epochs of homologous mesocorticolimbic nodes revealed similar connectivity. Altogether, these results identify specific connectivity conserved across species within the mesocorticolimbic circuit that may underlie mechanisms of NAc DBS.
Collapse
|
11
|
Beane CR, Lewis DG, Bruns Vi N, Pikus KL, Durfee MH, Zegarelli RA, Perry TW, Sandoval O, Radke AK. Cholinergic mu-opioid receptor deletion alters reward preference and aversion-resistance. Neuropharmacology 2024; 255:110019. [PMID: 38810926 DOI: 10.1016/j.neuropharm.2024.110019] [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: 12/15/2023] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
The endogenous opioid system has been implicated in alcohol consumption and preference in both humans and animals. The mu opioid receptor (MOR) is expressed on multiple cells in the striatum, however little is known about the contributions of specific MOR populations to alcohol drinking behaviors. The current study used mice with a genetic deletion of MOR in cholinergic cells (ChAT-Cre/Oprm1fl/fl) to examine the role of MORs expressed in cholinergic interneurons (CINs) in home cage self-administration paradigms. Male and female ChAT-Cre/Oprm1fl/fl mice were generated and heterozygous Cre+ (knockout) and Cre- (control) mice were tested for alcohol consumption in two drinking paradigms: limited access "Drinking in the Dark" and intermittent access. Quinine was added to the drinking bottles in the DID experiment to test aversion-resistant, "compulsive" drinking. Nicotine and sucrose drinking were also assessed so comparisons could be made with other rewarding substances. Cholinergic MOR deletion did not influence consumption or preference for ethanol (EtOH) in either drinking task. Differences were observed in aversion-resistance in males with Cre + mice tolerating lower concentrations of quinine than Cre-. In contrast to EtOH, preference for nicotine was reduced following cholinergic MOR deletion while sucrose consumption and preference was increased in Cre+ (vs. Cre-) females. Locomotor activity was also greater in females following the deletion. These results suggest that cholinergic MORs participate in preference for rewarding substances. Further, while they are not required for consumption of alcohol alone, cholinergic MORs may influence the tendency to drink despite negative consequences.
Collapse
Affiliation(s)
- Cambria R Beane
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Delainey G Lewis
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Nicolaus Bruns Vi
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Kat L Pikus
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Mary H Durfee
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Roman A Zegarelli
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Thomas W Perry
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Oscar Sandoval
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA
| | - Anna K Radke
- Department of Psychology and Center for Neuroscience and Behavior, Miami University, Oxford, OH, USA.
| |
Collapse
|
12
|
Xu Y, Lin Y, Yu M, Zhou K. The nucleus accumbens in reward and aversion processing: insights and implications. Front Behav Neurosci 2024; 18:1420028. [PMID: 39184934 PMCID: PMC11341389 DOI: 10.3389/fnbeh.2024.1420028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024] Open
Abstract
The nucleus accumbens (NAc), a central component of the brain's reward circuitry, has been implicated in a wide range of behaviors and emotional states. Emerging evidence, primarily drawing from recent rodent studies, suggests that the function of the NAc in reward and aversion processing is multifaceted. Prolonged stress or drug use induces maladaptive neuronal function in the NAc circuitry, which results in pathological conditions. This review aims to provide comprehensive and up-to-date insights on the role of the NAc in motivated behavior regulation and highlights areas that demand further in-depth analysis. It synthesizes the latest findings on how distinct NAc neuronal populations and pathways contribute to the processing of opposite valences. The review examines how a range of neuromodulators, especially monoamines, influence the NAc's control over various motivational states. Furthermore, it delves into the complex underlying mechanisms of psychiatric disorders such as addiction and depression and evaluates prospective interventions to restore NAc functionality.
Collapse
Affiliation(s)
| | | | | | - Kuikui Zhou
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
13
|
Ingebretson AE, Alonso-Caraballo Y, Razidlo JA, Lemos JC. Corticotropin releasing factor alters the functional diversity of accumbal cholinergic interneurons. J Neurophysiol 2024; 132:403-417. [PMID: 39106208 PMCID: PMC11427051 DOI: 10.1152/jn.00348.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 08/09/2024] Open
Abstract
Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum (DS), ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared with males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared with males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared with other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.NEW & NOTEWORTHY Cholinergic interneurons (ChIs) within the dorsal and ventral striatum can exert a major influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum, specifically the nucleus accumbens (NAc) shell, are understudied. Here, we report that NAc shell ChIs have heterogeneous ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide corticotropin releasing factor (CRF) and by the estrous cycle.
Collapse
Affiliation(s)
- Anna E Ingebretson
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - Yanaira Alonso-Caraballo
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - John A Razidlo
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| | - Julia C Lemos
- Department of Neuroscience, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
- Medical Discovery Team on Addiction, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
14
|
Marinescu AM, Labouesse MA. The nucleus accumbens shell: a neural hub at the interface of homeostatic and hedonic feeding. Front Neurosci 2024; 18:1437210. [PMID: 39139500 PMCID: PMC11319282 DOI: 10.3389/fnins.2024.1437210] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024] Open
Abstract
Feeding behavior is a complex physiological process regulated by the interplay between homeostatic and hedonic feeding circuits. Among the neural structures involved, the nucleus accumbens (NAc) has emerged as a pivotal region at the interface of these two circuits. The NAc comprises distinct subregions and in this review, we focus mainly on the NAc shell (NAcSh). Homeostatic feeding circuits, primarily found in the hypothalamus, ensure the organism's balance in energy and nutrient requirements. These circuits monitor peripheral signals, such as insulin, leptin, and ghrelin, and modulate satiety and hunger states. The NAcSh receives input from these homeostatic circuits, integrating information regarding the organism's metabolic needs. Conversely, so-called hedonic feeding circuits involve all other non-hunger and -satiety processes, i.e., the sensory information, associative learning, reward, motivation and pleasure associated with food consumption. The NAcSh is interconnected with hedonics-related structures like the ventral tegmental area and prefrontal cortex and plays a key role in encoding hedonic information related to palatable food seeking or consumption. In sum, the NAcSh acts as a crucial hub in feeding behavior, integrating signals from both homeostatic and hedonic circuits, to facilitate behavioral output via its downstream projections. Moreover, the NAcSh's involvement extends beyond simple integration, as it directly impacts actions related to food consumption. In this review, we first focus on delineating the inputs targeting the NAcSh; we then present NAcSh output projections to downstream structures. Finally we discuss how the NAcSh regulates feeding behavior and can be seen as a neural hub integrating homeostatic and hedonic feeding signals, via a functionally diverse set of projection neuron subpopulations.
Collapse
Affiliation(s)
- Alina-Măriuca Marinescu
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Marie A. Labouesse
- Brain, Wire and Behavior Group, Translational Nutritional Biology Laboratory, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
15
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. Curr Biol 2024; 34:3086-3101.e4. [PMID: 38925117 PMCID: PMC11279555 DOI: 10.1016/j.cub.2024.05.069] [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: 03/25/2024] [Revised: 04/25/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward rewards. Dopamine (DA) neurons in the ventral tegmental area (VTA) and substantia nigra (SNc) are crucial for this process, via engagement of a reciprocally connected network with their striatal targets. Critically, it remains unknown how dopamine neuron activity itself engages dopamine signals throughout the striatum, across learning. Here, we investigated how optogenetic Pavlovian cue conditioning of VTA or SNc dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. Although VTA stimulation-evoked robust dopamine release in NAc core, shell, and DMS, predictive cues preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS. Despite the development of vigorous movement late in training, conditioned dopamine signals did not emerge in the DLS, even during Pavlovian conditioning with SNc dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show a broad dissociation in the fundamental prediction and reward-related information generated by VTA and SNc dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale adaptations across the striatal network emerge during learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Val L Collins
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA
| | - Sonal Sinha
- Krieger School of Arts & Sciences, Johns Hopkins University, 3400 N. Charles St, Baltimore, MD 21218, USA
| | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA; Medical Discovery Team on Addiction, University of Minnesota, 2001 6th St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Zhang Y, Ma H, Bai Y, Hou X, Yang Y, Wang G, Li Y. Chronic Neuropathic Pain and Comorbid Depression Syndrome: From Neural Circuit Mechanisms to Treatment. ACS Chem Neurosci 2024; 15:2432-2444. [PMID: 38916052 DOI: 10.1021/acschemneuro.4c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024] Open
Abstract
Chronic neuropathic pain and comorbid depression syndrome (CDS) is a major worldwide health problem that affects the quality of life of patients and imposes a tremendous socioeconomic burden. More than half of patients with chronic neuropathic pain also suffer from moderate or severe depression. Due to the complex pathogenesis of CDS, there are no effective therapeutic drugs available. The lack of research on the neural circuit mechanisms of CDS limits the development of treatments. The purpose of this article is to provide an overview of the various circuits involved in CDS. Notably, activating some neural circuits can alleviate pain and/or depression, while activating other circuits can exacerbate these conditions. Moreover, we discuss current and emerging pharmacotherapies for CDS, such as ketamine. Understanding the circuit mechanisms of CDS may provide clues for the development of novel drug treatments for improved CDS management.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yafan Bai
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiaojuan Hou
- Hebei North University, Zhangjiakou, 075000, China
| | - Yixin Yang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Guyan Wang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, 100850, China
| |
Collapse
|
17
|
Hopf FW. Nucleus accumbens shell cholinergic interneurons potently drive binge alcohol drinking: A commentary on Sharma et al., 2024. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1243-1245. [PMID: 38811254 DOI: 10.1111/acer.15339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 05/31/2024]
Affiliation(s)
- Frederic Woodward Hopf
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
18
|
Ingebretson AE, Alonso-Caraballo Y, Razidlo JA, Lemos JC. Corticotropin releasing factor alters the functional diversity of accumbal cholinergic interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.17.558116. [PMID: 37745598 PMCID: PMC10516029 DOI: 10.1101/2023.09.17.558116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum, ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared to males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared to males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared to other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.
Collapse
|
19
|
Yuan S, Jiang SC, Zhang ZW, Li ZL, Hu J. Substance Addiction Rehabilitation Drugs. Pharmaceuticals (Basel) 2024; 17:615. [PMID: 38794185 PMCID: PMC11124501 DOI: 10.3390/ph17050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The relapse rate of substance abusers is high, and addiction rehabilitation adjunct drugs need to be developed urgently. There have been numerous reports on blocking the formation of substance addiction, but studies on drugs that can alleviate withdrawal symptoms are very limited. Both the dopamine transporter (DAT) hypothesis and D3 dopamine receptor (D3R) hypothesis are proposed. DAT activators reduce the extracellular dopamine level, and D3R antagonists reduce the neuron's sensitivity to dopamine, both of which may exacerbate the withdrawal symptoms subsequently. The D3R partial agonist SK608 has biased signaling properties via the G-protein-dependent pathway but did not induce D3R desensitization and, thus, may be a promising drug for the withdrawal symptoms. Drugs for serotoninergic neurons or GABAergic neurons and anti-inflammatory drugs may have auxiliary effects to addiction treatments. Drugs that promote structural synaptic plasticity are also discussed.
Collapse
Affiliation(s)
- Shu Yuan
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China;
| | - Si-Cong Jiang
- Haisco Pharmaceutical Group Comp. Ltd., Chengdu 611138, China;
| | - Zhong-Wei Zhang
- College of Resources, Sichuan Agricultural University, Chengdu 611130, China;
| | - Zi-Lin Li
- Department of Cardiovascular Surgery, Xijing Hospital, Medical University of the Air Force, Xi’an 710032, China;
| | - Jing Hu
- School of Medicine, Northwest University, Xi’an 710069, China;
| |
Collapse
|
20
|
Beane CR, Lewis DG, Bruns NK, Pikus KL, Durfee MH, Zegarelli RA, Perry TW, Sandoval O, Radke AK. Cholinergic mu-opioid receptor deletion alters reward preference and aversion-resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.13.566881. [PMID: 38014065 PMCID: PMC10680803 DOI: 10.1101/2023.11.13.566881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Heavy alcohol use and binge drinking are important contributors to alcohol use disorder (AUD). The endogenous opioid system has been implicated in alcohol consumption and preference in both humans and animals. The mu opioid receptor (MOR) is expressed on multiple cells in the striatum, however little is known about the contributions of specific MOR populations to alcohol drinking behaviors. The current study used mice with a genetic deletion of MOR in cholinergic cells (ChAT-Cre/Oprm1fl/fl) to examine the role of MORs expressed in cholinergic interneurons (CINs) in home cage self-administration paradigms. Male and female ChAT-Cre/Oprm1fl/fl mice were generated and heterozygous Cre+ (knockout) and Cre- (control) mice were tested for alcohol and nicotine consumption. In Experiment 1, binge-like and quinine-resistant drinking was tested using 15% ethanol (EtOH) in a two-bottle, limited-access Drinking in the Dark paradigm. Experiment 2 involved a six-week intermittent access paradigm in which mice received 20% EtOH, nicotine, and then a combination of the two drugs. Experiment 3 assessed locomotor activity, sucrose preference, and quinine sensitivity. Deleting MORs in cholinergic cells did not alter consumption of EtOH in Experiment 1 or 2. In Experiment 1, the MOR deletion resulted in greater consumption of quinine-adulterated EtOH in male Cre+ mice (vs. Cre-). In Experiment 2, Cre+ mice demonstrated a significantly lower preference for nicotine but did not differ from Cre- mice in nicotine or nicotine + EtOH consumption. Overall fluid consumption was also heightened in the Cre+ mice. In Experiment 3, Cre+ females were found to have greater locomotor activity and preference for sucrose vs. Cre- mice. These data suggest that cholinergic MORs are not required for EtOH, drinking behaviors but may contribute to aversion resistant EtOH drinking in a sex-dependent manner.
Collapse
|
21
|
Engel L, Wolff AR, Blake M, Collins VL, Sinha S, Saunders BT. Dopamine neurons drive spatiotemporally heterogeneous striatal dopamine signals during learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.01.547331. [PMID: 38585717 PMCID: PMC10996462 DOI: 10.1101/2023.07.01.547331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Environmental cues, through Pavlovian learning, become conditioned stimuli that invigorate and guide animals toward acquisition of rewards. Dopamine neurons in the ventral tegmental area (VTA) and substantia nigra (SNC) are crucial for this process. Dopamine neurons are embedded in a reciprocally connected network with their striatal targets, the functional organization of which remains poorly understood. Here, we investigated how learning during optogenetic Pavlovian cue conditioning of VTA or SNC dopamine neurons directs cue-evoked behavior and shapes subregion-specific striatal dopamine dynamics. We used a fluorescent dopamine biosensor to monitor dopamine in the nucleus accumbens (NAc) core and shell, dorsomedial striatum (DMS), and dorsolateral striatum (DLS). We demonstrate spatially heterogeneous, learning-dependent dopamine changes across striatal regions. While VTA stimulation evoked robust dopamine release in NAc core, shell, and DMS, cues predictive of this activation preferentially recruited dopamine release in NAc core, starting early in training, and DMS, late in training. Corresponding negative prediction error signals, reflecting a violation in the expectation of dopamine neuron activation, only emerged in the NAc core and DMS, and not the shell. Despite development of vigorous movement late in training, conditioned dopamine signals did not similarly emerge in the DLS, even during Pavlovian conditioning with SNC dopamine neuron activation, which elicited robust DLS dopamine release. Together, our studies show broad dissociation in the fundamental prediction and reward-related information generated by different dopamine neuron populations and signaled by dopamine across the striatum. Further, they offer new insight into how larger-scale plasticity across the striatal network emerges during Pavlovian learning to coordinate behavior.
Collapse
Affiliation(s)
- Liv Engel
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
- Current Address: Department of Psychology, University of Toronto
| | - Amy R Wolff
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Madelyn Blake
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | - Val L Collins
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| | | | - Benjamin T Saunders
- Department of Neuroscience, University of Minnesota
- Medical Discovery Team on Addiction, University of Minnesota
| |
Collapse
|
22
|
Hu Y, Du W, Qi J, Luo H, Zhang Z, Luo M, Wang Y. Comparative brain-wide mapping of ketamine- and isoflurane-activated nuclei and functional networks in the mouse brain. eLife 2024; 12:RP88420. [PMID: 38512722 PMCID: PMC10957177 DOI: 10.7554/elife.88420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024] Open
Abstract
Ketamine (KET) and isoflurane (ISO) are two widely used general anesthetics, yet their distinct and shared neurophysiological mechanisms remain elusive. In this study, we conducted a comparative analysis of the effects of KET and ISO on c-Fos expression across the mouse brain, utilizing hierarchical clustering and c-Fos-based functional network analysis to evaluate the responses of individual brain regions to each anesthetic. Our findings reveal that KET activates a wide range of brain regions, notably in the cortical and subcortical nuclei involved in sensory, motor, emotional, and reward processing, with the temporal association areas (TEa) as a strong hub, suggesting a top-down mechanism affecting consciousness by primarily targeting higher order cortical networks. In contrast, ISO predominantly influences brain regions in the hypothalamus, impacting neuroendocrine control, autonomic function, and homeostasis, with the locus coeruleus (LC) as a connector hub, indicating a bottom-up mechanism in anesthetic-induced unconsciousness. KET and ISO both activate brain areas involved in sensory processing, memory and cognition, reward and motivation, as well as autonomic and homeostatic control, highlighting their shared effects on various neural pathways. In conclusion, our results highlight the distinct but overlapping effects of KET and ISO, enriching our understanding of the mechanisms underlying general anesthesia.
Collapse
Affiliation(s)
- Yue Hu
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Wenjie Du
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Jiangtao Qi
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Huoqing Luo
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Mengqiang Luo
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
23
|
Wojick JA, Paranjapye A, Chiu JK, Mahmood M, Oswell C, Kimmey BA, Wooldridge LM, McCall NM, Han A, Ejoh LL, Chehimi SN, Crist RC, Reiner BC, Korb E, Corder G. A nociceptive amygdala-striatal pathway for chronic pain aversion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579947. [PMID: 38405972 PMCID: PMC10888915 DOI: 10.1101/2024.02.12.579947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The basolateral amygdala (BLA) is essential for assigning positive or negative valence to sensory stimuli. Noxious stimuli that cause pain are encoded by an ensemble of nociceptive BLA projection neurons (BLAnoci ensemble). However, the role of the BLAnoci ensemble in mediating behavior changes and the molecular signatures and downstream targets distinguishing this ensemble remain poorly understood. Here, we show that the same BLAnoci ensemble neurons are required for both acute and chronic neuropathic pain behavior. Using single nucleus RNA-sequencing, we characterized the effect of acute and chronic pain on the BLA and identified enrichment for genes with known functions in axonal and synaptic organization and pain perception. We thus examined the brain-wide targets of the BLAnoci ensemble and uncovered a previously undescribed nociceptive hotspot of the nucleus accumbens shell (NAcSh) that mirrors the stability and specificity of the BLAnoci ensemble and is recruited in chronic pain. Notably, BLAnoci ensemble axons transmit acute and neuropathic nociceptive information to the NAcSh, highlighting this nociceptive amygdala-striatal circuit as a unique pathway for affective-motivational responses across pain states.
Collapse
Affiliation(s)
- Jessica A. Wojick
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Alekh Paranjapye
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Juliann K. Chiu
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malaika Mahmood
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Corinna Oswell
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Blake A. Kimmey
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lisa M. Wooldridge
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nora M. McCall
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan Han
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lindsay L. Ejoh
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Samar Nasser Chehimi
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard C. Crist
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin C. Reiner
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erica Korb
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gregory Corder
- Dept. of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Dept. of Neuroscience, Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Fang LZ, Creed MC. Updating the striatal-pallidal wiring diagram. Nat Neurosci 2024; 27:15-27. [PMID: 38057614 DOI: 10.1038/s41593-023-01518-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/06/2023] [Indexed: 12/08/2023]
Abstract
The striatal and pallidal complexes are basal ganglia structures that orchestrate learning and execution of flexible behavior. Models of how the basal ganglia subserve these functions have evolved considerably, and the advent of optogenetic and molecular tools has shed light on the heterogeneity of subcircuits within these pathways. However, a synthesis of how molecularly diverse neurons integrate into existing models of basal ganglia function is lacking. Here, we provide an overview of the neurochemical and molecular diversity of striatal and pallidal neurons and synthesize recent circuit connectivity studies in rodents that takes this diversity into account. We also highlight anatomical organizational principles that distinguish the dorsal and ventral basal ganglia pathways in rodents. Future work integrating the molecular and anatomical properties of striatal and pallidal subpopulations may resolve controversies regarding basal ganglia network function.
Collapse
Affiliation(s)
- Lisa Z Fang
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland and Labrador, Canada
| | - Meaghan C Creed
- Washington University Pain Center, Department of Anesthesiology, St. Louis, MO, USA.
- Departments of Psychiatry, Neuroscience and Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
25
|
Cubello J, Marvin E, Conrad K, Merrill AK, George JV, Welle K, Jackson BP, Chalupa D, Oberdörster G, Sobolewski M, Cory-Slechta DA. The contributions of neonatal inhalation of copper to air pollution-induced neurodevelopmental outcomes in mice. Neurotoxicology 2024; 100:55-71. [PMID: 38081392 PMCID: PMC10842733 DOI: 10.1016/j.neuro.2023.12.007] [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] [Received: 10/05/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Exposures to ambient ultrafine particle (UFP) air pollution (AP) during the early postnatal period in mice (equivalent to human third trimester brain development) produce male-biased changes in brain structure, including ventriculomegaly, reduced brain myelination, alterations in neurotransmitters and glial activation, as well as impulsive-like behavioral characteristics, all of which are also features characteristic of male-biased neurodevelopmental disorders (NDDs). The purpose of this study was to ascertain the extent to which inhaled Cu, a common contaminant of AP that is also dysregulated across multiple NDDs, might contribute to these phenotypes. For this purpose, C57BL/6J mice were exposed from postnatal days 4-7 and 10-13 for 4 hr/day to inhaled copper oxide (CuxOy) nanoparticles at an environmentally relevant concentration averaging 171.9 ng/m3. Changes in brain metal homeostasis and neurotransmitter levels were determined following termination of exposure (postnatal day 14), while behavioral changes were assessed in adulthood. CuxOy inhalation modified cortical metal homeostasis and produced male-biased disruption of striatal neurotransmitters, with marked increases in dopaminergic function, as well as excitatory/inhibitory imbalance and reductions in serotonergic function. Impulsive-like behaviors in a fixed ratio (FR) waiting-for-reward schedule and a fixed interval (FI) schedule of food reward occurred in both sexes, but more prominently in males, effects which could not be attributed to altered locomotor activity or short-term memory. Inhaled Cu as from AP exposures, at environmentally relevant levels experienced during development, may contribute to impaired brain function, as shown by its ability to disrupt brain metal homeostasis and striatal neurotransmission. In addition, its ability to evoke impulsive-like behavior, particularly in male offspring, may be related to striatal dopaminergic dysfunction that is known to mediate such behaviors. As such, regulation of air Cu levels may be protective of public health.
Collapse
Affiliation(s)
- Janine Cubello
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Elena Marvin
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Katherine Conrad
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alyssa K Merrill
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jithin V George
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kevin Welle
- Proteomics Core, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - David Chalupa
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Günter Oberdörster
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
26
|
Belilos A, Gray C, Sanders C, Black D, Mays E, Richie C, Sengupta A, Hake H, Francis TC. Nucleus accumbens local circuit for cue-dependent aversive learning. Cell Rep 2023; 42:113488. [PMID: 37995189 PMCID: PMC10795009 DOI: 10.1016/j.celrep.2023.113488] [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: 03/17/2023] [Revised: 10/06/2023] [Accepted: 11/08/2023] [Indexed: 11/25/2023] Open
Abstract
Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient, which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We describe a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor-expressing projection neurons are required for cue-dependent fear learning. Additionally, we find that fear learning and recall is dependent on distinct projection neuron subtypes. Our work demonstrates a critical role for nucleus accumbens substance P in cue-dependent aversive learning.
Collapse
Affiliation(s)
- Andrew Belilos
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Cortez Gray
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Christie Sanders
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Destiny Black
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Elizabeth Mays
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | - Christopher Richie
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ayesha Sengupta
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Holly Hake
- Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - T Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
27
|
Zhou ZC, Gordon-Fennell A, Piantadosi SC, Ji N, Smith SL, Bruchas MR, Stuber GD. Deep-brain optical recording of neural dynamics during behavior. Neuron 2023; 111:3716-3738. [PMID: 37804833 PMCID: PMC10843303 DOI: 10.1016/j.neuron.2023.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 10/09/2023]
Abstract
In vivo fluorescence recording techniques have produced landmark discoveries in neuroscience, providing insight into how single cell and circuit-level computations mediate sensory processing and generate complex behaviors. While much attention has been given to recording from cortical brain regions, deep-brain fluorescence recording is more complex because it requires additional measures to gain optical access to harder to reach brain nuclei. Here we discuss detailed considerations and tradeoffs regarding deep-brain fluorescence recording techniques and provide a comprehensive guide for all major steps involved, from project planning to data analysis. The goal is to impart guidance for new and experienced investigators seeking to use in vivo deep fluorescence optical recordings in awake, behaving rodent models.
Collapse
Affiliation(s)
- Zhe Charles Zhou
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Adam Gordon-Fennell
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Sean C Piantadosi
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Na Ji
- Department of Physics, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Spencer LaVere Smith
- Department of Electrical and Computer Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Michael R Bruchas
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| | - Garret D Stuber
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA; Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
28
|
Sullere S, Kunczt A, McGehee DS. A cholinergic circuit that relieves pain despite opioid tolerance. Neuron 2023; 111:3414-3434.e15. [PMID: 37734381 PMCID: PMC10843525 DOI: 10.1016/j.neuron.2023.08.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/19/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023]
Abstract
Chronic pain is a tremendous burden for afflicted individuals and society. Although opioids effectively relieve pain, significant adverse outcomes limit their utility and efficacy. To investigate alternate pain control mechanisms, we explored cholinergic signaling in the ventrolateral periaqueductal gray (vlPAG), a critical nexus for descending pain modulation. Biosensor assays revealed that pain states decreased acetylcholine release in vlPAG. Activation of cholinergic projections from the pedunculopontine tegmentum to vlPAG relieved pain, even in opioid-tolerant conditions, through ⍺7 nicotinic acetylcholine receptors (nAChRs). Activating ⍺7 nAChRs with agonists or stimulating endogenous acetylcholine inhibited vlPAG neuronal activity through Ca2+ and peroxisome proliferator-activated receptor α (PPAR⍺)-dependent signaling. In vivo 2-photon imaging revealed that chronic pain induces aberrant excitability of vlPAG neuronal ensembles and that ⍺7 nAChR-mediated inhibition of these cells relieves pain, even after opioid tolerance. Finally, pain relief through these cholinergic mechanisms was not associated with tolerance, reward, or withdrawal symptoms, highlighting its potential clinical relevance.
Collapse
Affiliation(s)
- Shivang Sullere
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA
| | - Alissa Kunczt
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Daniel S McGehee
- Committee on Neurobiology, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia and Critical Care, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
29
|
Zell V, Teuns G, Needham AS, Mukherjee S, Roscoe N, Le M, Fourgeaud L, Woodruff G, Bhattacharya A, Marella M, Bonaventure P, Drevets WC, Balana B. Characterization of Selective M 5 Acetylcholine Muscarinic Receptor Modulators on Dopamine Signaling in the Striatum. J Pharmacol Exp Ther 2023; 387:226-234. [PMID: 37679045 DOI: 10.1124/jpet.123.001737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The type-5 muscarinic acetylcholine receptor (mAChR, M5) is almost exclusively expressed in dopamine (DA) neurons of the ventral tegmental area and substantia nigra pars compacta; therefore, they are ideally located to modulate DA signaling and underlying behaviors. However, the role of M5 in shaping DA release is still poorly characterized. In this study, we first quantitatively mapped the expression of M5 in different neurons of the mouse midbrain, then used voltammetry in mouse striatum to evaluate the effect of M5-selective modulators on DA release. The M5 negative allosteric modulator ML375 significantly decreased electrically evoked DA release and blocked the effect of Oxotremorine-M (Oxo-M; nonselective mAChR agonist) on DA release in the presence of an acetylcholine nicotinic receptor blocker. Conversely, the M5 positive allosteric modulator VU 0365114 significantly increased electrically evoked DA release and the Oxo-M effect on DA release. We then assessed M5's impact on mesolimbic circuit function in vivo. Although psychostimulant-induced locomotor activity models in knockout mice have previously been used to characterize the role of M5 in DA transmission, the results of these studies conflict, leading us to select a different in vivo model, namely a cocaine self-administration paradigm. In contrast to a previous study that also used this model, in the current study, administration of ML375 did not decrease cocaine self-administration in rats (using fixed and progressive ratio). These conflicting results illustrate the complexity of M5 modulation and the need to further characterize its involvement in the regulation of dopamine signaling, central to multiple neuropsychiatric diseases. SIGNIFICANCE STATEMENT: This work describes the type-5 muscarinic receptor (M5) pattern of expression within the midbrain as well as its physiological modulation by selective compounds at the axon terminal level in the striatum, where M5 directly shapes dopamine transmission. It offers the first direct readout of mesolimbic dopamine release modulation by M5, highlighting its role in regulating neurocircuits implicated in the pathophysiology of neuropsychiatric disorders such as substance use disorders, major depressive disorder, and schizophrenia.
Collapse
Affiliation(s)
- Vivien Zell
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Greetje Teuns
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Alexandra Stormy Needham
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Sruti Mukherjee
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Nathaniel Roscoe
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Michelle Le
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Lawrence Fourgeaud
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Grace Woodruff
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Anindya Bhattacharya
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Mathieu Marella
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Pascal Bonaventure
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Wayne C Drevets
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| | - Bartosz Balana
- Janssen Research and Development LLC, La Jolla, California (V.Z., A.S.N., S.M., N.R., M.L., L.F., G.W., A.B., M.M., P.B., W.C.D., B.B.) and Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium (G.T.)
| |
Collapse
|
30
|
Domingues AV, Rodrigues AJ, Soares-Cunha C. A novel perspective on the role of nucleus accumbens neurons in encoding associative learning. FEBS Lett 2023; 597:2601-2610. [PMID: 37643893 DOI: 10.1002/1873-3468.14727] [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: 05/31/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
The nucleus accumbens (NAc) has been considered a key brain region for encoding reward/aversion and cue-outcome associations. These processes are encoded by medium spiny neurons that express either dopamine receptor D1 (D1-MSNs) or D2 (D2-MSNs). Despite the well-established role of NAc neurons in encoding reward/aversion, the underlying processing by D1-/D2-MSNs remains largely unknown. Recent electrophysiological, optogenetic and calcium imaging studies provided insight on the complex role of D1- and D2-MSNs in these behaviours and helped to clarify their involvement in associative learning. Here, we critically discuss findings supporting an intricate and complementary role of NAc D1- and D2-MSNs in associative learning, emphasizing the need for additional studies in order to fully understand the role of these neurons in behaviour.
Collapse
Affiliation(s)
- Ana Verónica Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
31
|
Meng F, Wang L. Bidirectional mechanism of comorbidity of depression and insomnia based on synaptic plasticity. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1518-1528. [PMID: 38432881 PMCID: PMC10929903 DOI: 10.11817/j.issn.1672-7347.2023.230082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Indexed: 03/05/2024]
Abstract
Insomnia is one of the most common accompanying symptoms of depression, with both sharing highly overlapping molecular pathways. The same pathological changes can trigger comorbidity of insomnia and depression, which further forms a vicious cycle with the involvement of more mechanisms and disease progression. Thus, understanding the potential interaction mechanisms between insomnia and depression is critical for clinical diagnosis and treatment. Comorbidity genetic factors, the hypothalamic-pituitary-adrenal axis, along with circadian rhythms of cortisol and the brain reward mechanism, are important ways in contributing to the comorbidity occurrence and development. However, owing to lack of pertinent investigational data, intricate molecular mechanisms necessitate further elaboration. Synaptic plasticity is a solid foundation for neural homeostasis. Pathological alterations of depression and insomnia may perturb the production and release of neurotransmitter, dendritic spine remodeling and elimination, which converges and reflects in aberrant synaptic dynamics. Hence, the introduction of synaptic plasticity research route and the construction of a comprehensive model of depression and insomnia comorbidity can provide new ideas for clinical depression insomnia comorbidity treatment plans.
Collapse
Affiliation(s)
- Fanhao Meng
- First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin 150040.
| | - Long Wang
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
32
|
Wang A, Zhou Y, Chen H, Jin J, Mao Y, Tao S, Qiu T. Inhibition of SK Channels in VTA Affects Dopaminergic Neurons to Improve the Depression-Like Behaviors of Post-Stroke Depression Rats. Neuropsychiatr Dis Treat 2023; 19:2127-2139. [PMID: 37840624 PMCID: PMC10572402 DOI: 10.2147/ndt.s426091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023] Open
Abstract
Purpose This study aimed to investigate the effect of small-conductance calcium-activated potassium channels (SK channels) on the dopaminergic (DA) neuron pathways in the ventral tegmental area (VTA) during the pathogenesis of post-stroke depression (PSD) and explore the improvement of PSD by inhibiting the SK channels. Patients and Methods Four groups of Sprague-Dawley rats were randomly divided: Control, PSD, SK channel inhibitor (apamin) and SK channel activator (CyPPA) groups. In both control and CyPPA groups, sham surgery was performed. In the other two groups, middle cerebral arteries were occluded. The behavioral indicators related to depression in different groups were compared. Immunofluorescence was used to measure the activity of DA neurons in the VTA, while qRT-PCR was used to assess the expression of SK channel genes. Results The results showed that apamin treatment improved behavioral indicators related to depression compared to the PSD group. Furthermore, the qRT-PCR analysis revealed differential expression of the KCNN1 and KCNN3 subgenes of the SK channels in each group. Immunofluorescence analysis revealed an increase in the expression of DA neurons in the VTA of the PSD group, which was subsequently reduced upon apamin intervention. Conclusion This study suggests that SK channel activation following stroke contributes to depression-related behaviors in PSD rats through increased expression of DA neurons in the VTA. And depression-related behavior is improved in PSD rats by inhibiting the SK channels. The results of this study provide a new understanding of PSD pathogenesis and the possibility of developing new strategies to prevent PSD by targeting SK channels.
Collapse
Affiliation(s)
- Anqi Wang
- First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| | - Yujia Zhou
- Second Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| | - Huangying Chen
- First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| | - Jiawei Jin
- First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| | - Yingqi Mao
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, People’s Republic of China
| | - Shuiliang Tao
- Basic Medicine College, Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| | - Tao Qiu
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
33
|
Belilos A, Gray C, Sanders C, Black D, Mays E, Richie CT, Sengupta A, Hake HS, Francis TC. Nucleus Accumbens Local Circuit for Cue-Dependent Aversive Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527338. [PMID: 36798245 PMCID: PMC9934565 DOI: 10.1101/2023.02.06.527338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Response to threatening environmental stimuli requires detection and encoding of important environmental features that dictate threat. Aversive events are highly salient which promotes associative learning about stimuli that signal this threat. The nucleus accumbens is uniquely positioned to process this salient, aversive information and promote motivated output, through plasticity on the major projection neurons in the brain area. We uncovered a nucleus accumbens core local circuit whereby excitatory plasticity facilitates learning and recall of discrete aversive cues. We demonstrate that putative nucleus accumbens substance P release and long-term excitatory plasticity on dopamine 2 receptor expressing projection neurons is required for cue-dependent fear learning. Additionally, we found fear learning and recall were dependent on distinct projection-neuron subtypes. Our work demonstrates a critical role for Nucleus Accumbens substance P in cue-dependent aversive learning.
Collapse
|
34
|
Toth BA, Chang KS, Fechtali S, Burgess CR. Dopamine release in the nucleus accumbens promotes REM sleep and cataplexy. iScience 2023; 26:107613. [PMID: 37664637 PMCID: PMC10470413 DOI: 10.1016/j.isci.2023.107613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023] Open
Abstract
Patients with the sleep disorder narcolepsy suffer from excessive daytime sleepiness, disrupted nighttime sleep, and cataplexy-the abrupt loss of postural muscle tone during wakefulness, often triggered by strong emotion. The dopamine (DA) system is implicated in both sleep-wake states and cataplexy, but little is known about the function of DA release in the striatum and sleep disorders. Recording DA release in the ventral striatum revealed orexin-independent changes across sleep-wake states as well as striking increases in DA release in the ventral, but not dorsal, striatum prior to cataplexy onset. Tonic low-frequency stimulation of ventral tegmental efferents in the ventral striatum suppressed both cataplexy and rapid eye movement (REM) sleep, while phasic high-frequency stimulation increased cataplexy propensity and decreased the latency to REM sleep. Together, our findings demonstrate a functional role of DA release in the striatum in regulating cataplexy and REM sleep.
Collapse
Affiliation(s)
- Brandon A. Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Katie S. Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Fechtali
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
35
|
Chantranupong L, Beron CC, Zimmer JA, Wen MJ, Wang W, Sabatini BL. Dopamine and glutamate regulate striatal acetylcholine in decision-making. Nature 2023; 621:577-585. [PMID: 37557915 PMCID: PMC10511323 DOI: 10.1038/s41586-023-06492-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Striatal dopamine and acetylcholine are essential for the selection and reinforcement of motor actions and decision-making1. In vitro studies have revealed an intrastriatal circuit in which acetylcholine, released by cholinergic interneurons (CINs), drives the release of dopamine, and dopamine, in turn, inhibits the activity of CINs through dopamine D2 receptors (D2Rs). Whether and how this circuit contributes to striatal function in vivo is largely unknown. Here, to define the role of this circuit in a living system, we monitored acetylcholine and dopamine signals in the ventrolateral striatum of mice performing a reward-based decision-making task. We establish that dopamine and acetylcholine exhibit multiphasic and anticorrelated transients that are modulated by decision history and reward outcome. Dopamine dynamics and reward encoding do not require the release of acetylcholine by CINs. However, dopamine inhibits acetylcholine transients in a D2R-dependent manner, and loss of this regulation impairs decision-making. To determine how other striatal inputs shape acetylcholine signals, we assessed the contribution of cortical and thalamic projections, and found that glutamate release from both sources is required for acetylcholine release. Altogether, we uncover a dynamic relationship between dopamine and acetylcholine during decision-making, and reveal multiple modes of CIN regulation. These findings deepen our understanding of the neurochemical basis of decision-making and behaviour.
Collapse
Affiliation(s)
- Lynne Chantranupong
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Celia C Beron
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Joshua A Zimmer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Michelle J Wen
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Wengang Wang
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, USA.
| |
Collapse
|
36
|
Bouarab C, Wynalda M, Thompson BV, Khurana A, Cody CR, Kisner A, Polter AM. Sex-specific adaptations to VTA circuits following subchronic stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551665. [PMID: 37577542 PMCID: PMC10418168 DOI: 10.1101/2023.08.02.551665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Dysregulation of the mesolimbic reward circuitry is implicated in the pathophysiology of stress-related illnesses such as depression and anxiety. These disorders are more frequently diagnosed in females, and sex differences in the response to stress are likely to be one factor that leads to enhanced vulnerability of females. In this study, we use subchronic variable stress (SCVS), a model in which females are uniquely vulnerable to behavioral disturbances, to investigate sexually divergent mechanisms of regulation of the ventral tegmental area by stress. Using slice electrophysiology, we find that female, but not male mice have a reduction in the ex vivo firing rate of VTA dopaminergic neurons following SCVS. Surprisingly, both male and female animals show an increase in inhibitory tone onto VTA dopaminergic neurons and an increase in the firing rate of VTA GABAergic neurons. In males, however, this is accompanied by a robust increase in excitatory synaptic tone onto VTA dopamine neurons. This supports a model by which SCVS recruits VTA GABA neurons to inhibit dopaminergic neurons in both male and female mice, but males are protected from diminished functioning of the dopaminergic system by a compensatory upregulation of excitatory synapses.
Collapse
Affiliation(s)
- Chloé Bouarab
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Institut Pasteur, 25-28 rue du Docteur Roux, 75015 Paris
| | - Megan Wynalda
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Brittney V. Thompson
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Department of Psychology, Florida State University, Tallahasse, FL, 32306
| | - Ambika Khurana
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Caitlyn R. Cody
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Department of Psychology, Northeastern University, Boston, MA, 02115
| | - Alexandre Kisner
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
- Current address: Department of Neuroscience, American University, Washington DC 20016
| | - Abigail M. Polter
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| |
Collapse
|
37
|
Han Z, Luo N, Ma W, Liu X, Cai Y, Kou J, Wang J, Li L, Peng S, Xu Z, Zhang W, Qiu Y, Wu Y, Ye C, Lin K, Xu F. AAV11 enables efficient retrograde targeting of projection neurons and enhances astrocyte-directed transduction. Nat Commun 2023; 14:3792. [PMID: 37365155 DOI: 10.1038/s41467-023-39554-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
Viral tracers that enable efficient retrograde labeling of projection neurons are powerful vehicles for structural and functional dissections of the neural circuit and for the treatment of brain diseases. Currently, some recombinant adeno-associated viruses (rAAVs) based on capsid engineering are widely used for retrograde tracing, but display undesirable brain area selectivity due to inefficient retrograde transduction in certain neural connections. Here we developed an easily editable toolkit to produce high titer AAV11 and demonstrated that it exhibits potent and stringent retrograde labeling of projection neurons in adult male wild-type or Cre transgenic mice. AAV11 can function as a powerful retrograde viral tracer complementary to AAV2-retro in multiple neural connections. In combination with fiber photometry, AAV11 can be used to monitor neuronal activities in the functional network by retrograde delivering calcium-sensitive indicator under the control of a neuron-specific promoter or the Cre-lox system. Furthermore, we showed that GfaABC1D promoter embedding AAV11 is superior to AAV8 and AAV5 in astrocytic tropism in vivo, combined with bidirectional multi-vector axoastrocytic labeling, AAV11 can be used to study neuron-astrocyte connection. Finally, we showed that AAV11 allows for analyzing circuit connectivity difference in the brains of the Alzheimer's disease and control mice. These properties make AAV11 a promising tool for mapping and manipulating neural circuits and for gene therapy of some neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Zengpeng Han
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Nengsong Luo
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Wenyu Ma
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Yuxiang Cai
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China
| | - Jiaxin Kou
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Lei Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
| | - Siqi Peng
- College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Zihong Xu
- College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Wen Zhang
- College of Life Sciences, Wuhan University, Wuhan, 430072, PR China
| | - Yuxiang Qiu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Yang Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Chaohui Ye
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China
- University of Chinese Academy of Sciences, 100049, Beijing, PR China
| | - Kunzhang Lin
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
| | - Fuqiang Xu
- Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, PR China.
- Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, PR China.
- University of Chinese Academy of Sciences, 100049, Beijing, PR China.
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, PR China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, PR China.
| |
Collapse
|
38
|
Mohebi A, Collins VL, Berke JD. Accumbens cholinergic interneurons dynamically promote dopamine release and enable motivation. eLife 2023; 12:e85011. [PMID: 37272423 PMCID: PMC10259987 DOI: 10.7554/elife.85011] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 05/30/2023] [Indexed: 06/06/2023] Open
Abstract
Motivation to work for potential rewards is critically dependent on dopamine (DA) in the nucleus accumbens (NAc). DA release from NAc axons can be controlled by at least two distinct mechanisms: (1) action potentials propagating from DA cell bodies in the ventral tegmental area (VTA), and (2) activation of β2* nicotinic receptors by local cholinergic interneurons (CINs). How CIN activity contributes to NAc DA dynamics in behaving animals is not well understood. We monitored DA release in the NAc Core of awake, unrestrained rats using the DA sensor RdLight1, while simultaneously monitoring or manipulating CIN activity at the same location. CIN stimulation rapidly evoked DA release, and in contrast to slice preparations, this DA release showed no indication of short-term depression or receptor desensitization. The sound of unexpected food delivery evoked a brief joint increase in CIN population activity and DA release, with a second joint increase as rats approached the food. In an operant task, we observed fast ramps in CIN activity during approach behaviors, either to start the trial or to collect rewards. These CIN ramps co-occurred with DA release ramps, without corresponding changes in the firing of lateral VTA DA neurons. Finally, we examined the effects of blocking CIN influence over DA release through local NAc infusion of DHβE, a selective antagonist of β2* nicotinic receptors. DHβE dose-dependently interfered with motivated approach decisions, mimicking the effects of a DA antagonist. Our results support a key influence of CINs over motivated behavior via the local regulation of DA release.
Collapse
Affiliation(s)
- Ali Mohebi
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Val L Collins
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
| | - Joshua D Berke
- Department of Neurology, University of California, San FranciscoSan FranciscoUnited States
- Department of Psychiatry and Behavioral Sciences, University of California, San FranciscoSan FranciscoUnited States
- Neuroscience Graduate Program, University of California, San FranciscoSan FranciscoUnited States
- Weill Institute for Neurosciences, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
39
|
Toth BA, Chang KS, Burgess CR. Striatal dopamine regulates sleep states and narcolepsy-cataplexy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542872. [PMID: 37397994 PMCID: PMC10312558 DOI: 10.1101/2023.05.30.542872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Disruptions to sleep can be debilitating and have a severe effect on daily life. Patients with the sleep disorder narcolepsy suffer from excessive daytime sleepiness, disrupted nighttime sleep, and cataplexy - the abrupt loss of postural muscle tone (atonia) during wakefulness, often triggered by strong emotion. The dopamine (DA) system is implicated in both sleep-wake states and cataplexy, but little is known about the function of DA release in the striatum - a major output region of midbrain DA neurons - and sleep disorders. To better characterize the function and pattern of DA release in sleepiness and cataplexy, we combined optogenetics, fiber photometry, and sleep recordings in a murine model of narcolepsy (orexin-/-; OX KO) and in wildtype mice. Recording DA release in the ventral striatum revealed OX-independent changes across sleep-wake states as well as striking increases in DA release in the ventral, but not dorsal, striatum prior to cataplexy onset. Tonic low frequency stimulation of ventral tegmental efferents in the ventral striatum suppressed both cataplexy and REM sleep, while phasic high frequency stimulation increased cataplexy propensity and decreased the latency to rapid eye movement (REM) sleep. Together, our findings demonstrate a functional role of DA release in the striatum in regulating cataplexy and REM sleep.
Collapse
Affiliation(s)
- Brandon A. Toth
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
| | - Katie S. Chang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Christian R. Burgess
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
40
|
Jin R, Sun S, Hu Y, Zhang H, Sun X. Neuropeptides Modulate Feeding via the Dopamine Reward Pathway. Neurochem Res 2023:10.1007/s11064-023-03954-4. [PMID: 37233918 DOI: 10.1007/s11064-023-03954-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Dopamine (DA) is a catecholamine neurotransmitter widely distributed in the central nervous system. It participates in various physiological functions, such as feeding, anxiety, fear, sleeping and arousal. The regulation of feeding is exceptionally complex, involving energy homeostasis and reward motivation. The reward system comprises the ventral tegmental area (VTA), nucleus accumbens (NAc), hypothalamus, and limbic system. This paper illustrates the detailed mechanisms of eight typical orexigenic and anorexic neuropeptides that regulate food intake through the reward system. According to recent literature, neuropeptides released from the hypothalamus and other brain regions regulate reward feeding predominantly through dopaminergic neurons projecting from the VTA to the NAc. In addition, their effect on the dopaminergic system is mediated by the prefrontal cortex, paraventricular thalamus, laterodorsal tegmental area, amygdala, and complex neural circuits. Research on neuropeptides involved in reward feeding can help identify more targets to treat diseases with metabolic disorders, such as obesity.
Collapse
Affiliation(s)
- Ruijie Jin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Shanbin Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Yang Hu
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Hongfei Zhang
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
- Department of Clinical Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
41
|
Petrican R, Fornito A. Adolescent neurodevelopment and psychopathology: The interplay between adversity exposure and genetic risk for accelerated brain ageing. Dev Cogn Neurosci 2023; 60:101229. [PMID: 36947895 PMCID: PMC10041470 DOI: 10.1016/j.dcn.2023.101229] [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: 12/08/2022] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
In adulthood, stress exposure and genetic risk heighten psychological vulnerability by accelerating neurobiological senescence. To investigate whether molecular and brain network maturation processes play a similar role in adolescence, we analysed genetic, as well as longitudinal task neuroimaging (inhibitory control, incentive processing) and early life adversity (i.e., material deprivation, violence) data from the Adolescent Brain and Cognitive Development study (N = 980, age range: 9-13 years). Genetic risk was estimated separately for Major Depressive Disorder (MDD) and Alzheimer's Disease (AD), two pathologies linked to stress exposure and allegedly sharing a causal connection (MDD-to-AD). Adversity and genetic risk for MDD/AD jointly predicted functional network segregation patterns suggestive of accelerated (GABA-linked) visual/attentional, but delayed (dopamine [D2]/glutamate [GLU5R]-linked) somatomotor/association system development. A positive relationship between brain maturation and psychopathology emerged only among the less vulnerable adolescents, thereby implying that normatively maladaptive neurodevelopmental alterations could foster adjustment among the more exposed and genetically more stress susceptible youths. Transcriptomic analyses suggested that sensitivity to stress may underpin the joint neurodevelopmental effect of adversity and genetic risk for MDD/AD, in line with the proposed role of negative emotionality as a precursor to AD, likely to account for the alleged causal impact of MDD on dementia onset.
Collapse
Affiliation(s)
- Raluca Petrican
- Institute of Population Health, Department of Psychology, University of Liverpool, Bedford Street South, Liverpool L69 7ZA, United Kingdom.
| | - Alex Fornito
- The Turner Institute for Brain and Mental Health, School of Psychological Sciences, and Monash Biomedical Imaging, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
42
|
Olejniczak I, Begemann K, Wilhelm I, Oster H. The circadian neurobiology of reward. Acta Physiol (Oxf) 2023; 237:e13928. [PMID: 36625310 DOI: 10.1111/apha.13928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/29/2022] [Accepted: 01/02/2023] [Indexed: 01/11/2023]
Abstract
Circadian clocks are important regulators of physiology and behavior. In the brain, circadian clocks have been described in many centers of the central reward system. They affect neurotransmitter signaling, neuroendocrine circuits, and the sensitivity to external stimulation. Circadian disruption affects reward signaling, promoting the development of behavioral and substance use disorders. In this review, we summarize our current knowledge of circadian clock-reward crosstalk. We show how chronodisruption affects reward signaling in different animal models. We then translate these findings to circadian aspects of human reward (dys-) function and its clinical implications. Finally, we devise approaches to and challenges in implementing the concepts of circadian medicine in the therapy of substance use disorders.
Collapse
Affiliation(s)
- Iwona Olejniczak
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Kimberly Begemann
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| | - Ines Wilhelm
- Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany.,Translational Psychiatry Unit, Department of Psychiatry and Psychotherapy, University of Lübeck, Lübeck, Germany
| | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany.,Center of Brain, Behavior, and Metabolism, University of Lübeck, Lübeck, Germany
| |
Collapse
|
43
|
Fanni G, Kagios C, Roman E, Sundbom M, Wikström J, Haller S, Eriksson JW. Effects of gastric bypass surgery on brain connectivity responses to hypoglycemia. Endocrine 2023; 79:304-312. [PMID: 36459336 PMCID: PMC9892147 DOI: 10.1007/s12020-022-03253-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022]
Abstract
INTRODUCTION Roux-en-Y gastric bypass (RYGB) leads to beneficial effects on glucose homeostasis, and attenuated hormonal counterregulatory responses to hypoglycemia are likely to contribute. RYGB also induces alterations in neural activity of cortical and subcortical brain regions. We aimed to characterize RYGB-induced changes in resting-state connectivity of specific brain regions of interest for energy homeostasis and behavioral control during hypoglycemia. METHOD Ten patients with BMI > 35 kg/m2 were investigated with brain PET/MR imaging during a hyperinsulinemic normo- and hypoglycemic clamp, before and 4 months after RYGB. Hormonal levels were assessed throughout the clamp. Resting-state (RS) fMRI scans were acquired in the glucose-lowering phase of the clamp, and they were analyzed with a seed-to-voxel approach. RESULTS RS connectivity during initiation of hypoglycemia was significantly altered after RYGB between nucleus accumbens, thalamus, caudate, hypothalamus and their crosstalk with cortical and subcortical regions. Connectivity between the nucleus accumbens and the frontal pole was increased after RYGB, and this was associated with a reduction of ACTH (r = -0.639, p = 0.047) and cortisol (r = -0.635, p = 0.048) responses. Instead, connectivity between the caudate and the frontal pole after RYGB was reduced and this was associated with less attenuation of glucagon response during the hypoglycemic clamp (r = -0.728, p = 0.017), smaller reduction in fasting glucose (r = -0.798, p = 0.007) and less excess weight loss (r = 0.753, p = 0.012). No other significant associations were found between post-RYGB changes in ROI-to-voxel regional connectivity hormonal responses and metabolic or anthropometric outcomes. CONCLUSION RYGB alters brain connectivity during hypoglycemia of several neural pathways involved in reward, inhibitory control, and energy homeostasis. These changes are associated with altered hormonal responses to hypoglycemia and may be involved in the glucometabolic outcome of RYGB.
Collapse
Affiliation(s)
- Giovanni Fanni
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Christakis Kagios
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Erika Roman
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
- Department of Anatomy, Physiology and Biochemistry, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Magnus Sundbom
- Department of Surgical Sciences, Surgery, Uppsala University, Uppsala, Sweden
| | - Johan Wikström
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Uppsala, Sweden
| | - Sven Haller
- Department of Surgical Sciences, Neuroradiology, Uppsala University, Uppsala, Sweden
- CIMC-Centre d'Imagerie Médicale de Cornavin, Geneva, Switzerland
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
44
|
Hornung RS, Kinchington PR, Umorin M, Kramer PR. PAQR8 and PAQR9 expression is altered in the ventral tegmental area of aged rats infected with varicella zoster virus. Mol Pain 2023; 19:17448069231202598. [PMID: 37699860 PMCID: PMC10515525 DOI: 10.1177/17448069231202598] [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: 07/06/2023] [Revised: 08/17/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Infection with varicella zoster virus (VZV) results in chicken pox and reactivation of VZV results in herpes zoster (HZ) or what is often referred to as shingles. Patients with HZ experience decreased motivation and increased emotional distress consistent with functions of the ventral tegmental area (VTA) of the brain. In addition, activity within the ventral tegmental area is altered in patients with HZ. HZ primarily affects individuals that are older and the VTA changes with age. To begin to determine if the VTA has a role in HZ symptoms, a screen of 10,000 genes within the VTA in young and old male rats was completed after injecting the whisker pad with VZV. The two genes that had maximal change were membrane progesterone receptors PAQR8 (mPRβ) and PAQR9 (mPRε). Neurons and non-neuronal cells expressed both PAQR8 and PAQR9. PAQR8 and PAQR9 protein expression was significantly reduced after VZV injection of young males. In old rats PAQR9 protein expression was significantly increased after VZV injection and PAQR9 protein expression was reduced in aged male rats versus young rats. Consistent with previous results, pain significantly increased after VZV injection of the whisker pad and aged animals showed significantly more pain than young animals. Our data suggests that PAQR8 and PAQR9 expression is altered by VZV injection and that these changes are affected by age.
Collapse
Affiliation(s)
- Rebecca S Hornung
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Paul R Kinchington
- Department of Ophthalmology and of Molecular Microbiology and Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mikhail Umorin
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| | - Phillip R Kramer
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, TX, USA
| |
Collapse
|
45
|
Feng L, Lo H, You H, Wu W, Cheng X, Xin J, Ye Z, Chen X, Pan X. Loss of cannabinoid receptor 2 promotes α-Synuclein-induced microglial synaptic pruning in nucleus accumbens by modulating the pCREB-c-Fos signaling pathway and complement system. Exp Neurol 2023; 359:114230. [PMID: 36162511 DOI: 10.1016/j.expneurol.2022.114230] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/31/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022]
Abstract
The disruption of nucleus accumbens (NAc) function impacts mood and learning behavior in α-Synucleinopathy, in which microglial synaptic pruning plays a pivotal role in modulating the neuropathologic progression. Available literature documents that in microglia, the activation of cannabinoid receptor 2 (CB2R) decreases inflammation, but it remains obscured regarding the roles of CB2R in microglia-mediated synaptic pruning in the NAc during the neuropathological progression of α-Synucleinopathy. We adopted the fibrillar α-Synuclein (α-Syn) treatment to characterize the effect of genetic CB2R deletion on microglial function and the signaling pathway. CB2R knockout (CB2-/-) mice and wild-type (CB2+/+) mice were divided into the α-Syn or saline treatment groups. Biochemical and microscopy approaches, including immunofluorescence, real-time PCR, and western blotting, were employed to assess the changes in homeostasis of synaptic pruning in NAc under the α-Syn-induced microglia. Moreover, the underlying mechanisms of CB2R on α-Syn induced microglial activity was assessed in vitro. After the injection of α-Syn into the NAc, distinct microglial morphological changes and M1 phenotype transformation were observed between CB2-/- and CB2+/+ mice. Meanwhile, after the α-Syn treatment, CB2-/- mice showed an increased upregulation of CD68 protein and IL-1β mRNA but decreased brain-derived neurotrophic factor (BDNF) and TGF-β mRNA compared with CB2+/+ mice. Additionally, CB2-/- microglia after the treatment showed a highly enriched complement 3a receptor (C3aR) producing excessive pruning of cholinergic synapses but less engulfment of dopaminergic synapses. Mechanistically, the loss of CB2R function in the α-Syn stimulation triggered c-Fos activation in microglia, but not in neurons. Further inhibition of microglial CB2R functions under α-Syn stimulation activated the phosphorylated cAMP-response element-binding protein (pCREB)-c-Fos, which was closely related to the C3aR upregulation. Our results reveal a critical and mechanistic role of CB2R in altering the microglial function and its value in the homeostasis of synaptic circuits in the NAc under the α-Syn pathology.
Collapse
Affiliation(s)
- Linjuan Feng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Hsuan Lo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hanlin You
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Wei Wu
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaojuan Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Jiawei Xin
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China
| | - Zucheng Ye
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China
| | - Xiaochun Chen
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou 350001, China; Institute of Clinical Neurology, Fujian Medical University, 29 Xinquan Road, Fuzhou 350001, China; Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, 88 Jiaotong Road, Fuzhou 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, 1 Xueyuan Road, Fuzhou 350001, China.
| |
Collapse
|
46
|
Novel role of AMPK in cocaine reinforcement via regulating CRTC1. Transl Psychiatry 2022; 12:530. [PMID: 36587026 PMCID: PMC9805446 DOI: 10.1038/s41398-022-02299-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 01/01/2023] Open
Abstract
Repeated cocaine exposure causes compensatory neuroadaptations in neurons in the nucleus accumbens (NAc), a region that mediates reinforcing effects of drugs. Previous studies suggested a role for adenosine monophosphate-activated protein kinase (AMPK), a cellular energy sensor, in modulating neuronal morphology and membrane excitability. However, the potential involvement of AMPK in cocaine use disorder is still unclear. The present study employed a cocaine self-administration model in rats to investigate the effect of AMPK and its target cyclic adenosine monophosphate response element binding protein-regulated transcriptional co-activator 1 (CRTC1) on cocaine reinforcement and the motivation for cocaine. We found that intravenous cocaine self-administration significantly decreased AMPK activity in the NAc shell (NAcsh), which persisted for at least 7 days of withdrawal. Cocaine reinforcement, reflected by self-administration behavior, was significantly prevented or enhanced by augmenting or suppressing AMPK activity pharmacologically and genetically, respectively. No difference in sucrose self-administration behavior was found after the same manipulations. The inhibition of AMPK activity in the NAcsh also increased the motivation for cocaine in progressive-ratio schedules of reinforcement, whereas the activation of AMPK had no effect. The knockdown of CRTC1 in the NAcsh significantly impaired cocaine reinforcement, which was rescued by pharmacologically increasing AMPK activity. Altogether, these results indicate that AMPK in the NAcsh is critical for cocaine reinforcement, possibly via the regulation of CRTC1 signaling. These findings may help reveal potential therapeutic targets and have important implications for the treatment of cocaine use disorder and relapse.
Collapse
|
47
|
The antidepressant effect of nucleus accumbens deep brain stimulation is mediated by parvalbumin-positive interneurons in the dorsal dentate gyrus. Neurobiol Stress 2022; 21:100492. [DOI: 10.1016/j.ynstr.2022.100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/02/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
|
48
|
Zhou WL, Kim K, Ali F, Pittenger ST, Calarco CA, Mineur YS, Ramakrishnan C, Deisseroth K, Kwan AC, Picciotto MR. Activity of a direct VTA to ventral pallidum GABA pathway encodes unconditioned reward value and sustains motivation for reward. SCIENCE ADVANCES 2022; 8:eabm5217. [PMID: 36260661 PMCID: PMC9581470 DOI: 10.1126/sciadv.abm5217] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/01/2022] [Indexed: 05/28/2023]
Abstract
Dopamine signaling from the ventral tegmental area (VTA) plays critical roles in reward-related behaviors, but less is known about the functions of neighboring VTA GABAergic neurons. We show here that a primary target of VTA GABA projection neurons is the ventral pallidum (VP). Activity of VTA-to-VP-projecting GABA neurons correlates consistently with size and palatability of the reward and does not change following cue learning, providing a direct measure of reward value. Chemogenetic stimulation of this GABA projection increased activity of a subset of VP neurons that were active while mice were seeking reward. Optogenetic stimulation of this pathway improved performance in a cue-reward task and maintained motivation to work for reward over days. This VTA GABA projection provides information about reward value directly to the VP, likely distinct from the prediction error signal carried by VTA dopamine neurons.
Collapse
Affiliation(s)
- Wen-Liang Zhou
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Kristen Kim
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Farhan Ali
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Steven T. Pittenger
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Cali A. Calarco
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yann S. Mineur
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Alex C. Kwan
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
| | - Marina R. Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, New Haven, CT 06508, USA
- Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
49
|
Baimel C, Jang E, Scudder SL, Manoocheri K, Carter AG. Hippocampal-evoked inhibition of cholinergic interneurons in the nucleus accumbens. Cell Rep 2022; 40:111042. [PMID: 35793623 PMCID: PMC9302453 DOI: 10.1016/j.celrep.2022.111042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/12/2022] [Accepted: 06/12/2022] [Indexed: 02/04/2023] Open
Abstract
Cholinergic interneurons (ChIs) in the nucleus accumbens (NAc) play a central role in motivated behaviors and associated disorders. However, while the activation of ChIs has been well studied in the dorsal striatum, little is known about how they are engaged in the NAc. Here, we find that the ventral hippocampus (vHPC) and the paraventricular nucleus of the thalamus (PVT) are the main excitatory inputs to ChIs in the NAc medial shell. While the PVT activates ChIs, the vHPC evokes a pronounced pause in firing through prominent feedforward inhibition. In contrast to the dorsal striatum, this inhibition reflects strong connections onto ChIs from local parvalbumin interneurons. Our results reveal the mechanisms by which different long-range inputs engage ChIs, highlighting fundamental differences in local connectivity across the striatum.
Collapse
Affiliation(s)
- Corey Baimel
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Emily Jang
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Samantha L Scudder
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Kasra Manoocheri
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
50
|
Heshmati M, Bruchas MR. Historical and Modern Evidence for the Role of Reward Circuitry in Emergence. Anesthesiology 2022; 136:997-1014. [PMID: 35362070 PMCID: PMC9467375 DOI: 10.1097/aln.0000000000004148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Increasing evidence supports a role for brain reward circuitry in modulating arousal along with emergence from anesthesia. Emergence remains an important frontier for investigation, since no drug exists in clinical practice to initiate rapid and smooth emergence. This review discusses clinical and preclinical evidence indicating a role for two brain regions classically considered integral components of the mesolimbic brain reward circuitry, the ventral tegmental area and the nucleus accumbens, in emergence from propofol and volatile anesthesia. Then there is a description of modern systems neuroscience approaches to neural circuit investigations that will help span the large gap between preclinical and clinical investigation with the shared aim of developing therapies to promote rapid emergence without agitation or delirium. This article proposes that neuroscientists include models of whole-brain network activity in future studies to inform the translational value of preclinical investigations and foster productive dialogues with clinician anesthesiologists.
Collapse
Affiliation(s)
- Mitra Heshmati
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Biological Structure, University of Washington, Seattle, Washington
| | - Michael R Bruchas
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, and Department of Pharmacology, University of Washington, Seattle, Washington
| |
Collapse
|