1
|
Wu X, Tu M, Yu Z, Cao Z, Qu S, Chen N, Jin J, Xiong S, Yang J, Pei S, Xu M, Wang J, Shi Y, Gao L, Xie J, Li X, Fang J, Shao X. The efficacy and cerebral mechanism of intradermal acupuncture for major depressive disorder: a multicenter randomized controlled trial. Neuropsychopharmacology 2025; 50:1075-1083. [PMID: 39648209 DOI: 10.1038/s41386-024-02036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/05/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
New combinations or alternative therapies for major depressive disorder (MDD) are necessary. Intradermal acupuncture (IA) shows promise but requires further investigation regarding its efficacy, safety, and mechanisms. Conducted across 3 centers from November 2022 to January 2024, our randomized controlled trial included 120 participants with moderate to severe MDD, divided into the selective serotonin reuptake inhibitors (SSRIs), SSRIs plus sham IA (SSRIs + SIA), and SSRIs plus active IA (SSRIs + AIA) groups. Acupuncture groups received 10 sessions over 6 weeks at Shenmen (HT7), Neiguan (PC6), Sanyinjiao (SP6) and Taichong (LR3) bilaterally, followed by a 4-week follow-up. The primary outcome was changes in Hamilton Depression Rating Scale-17 (HAMD-17) scores at week 6. Furthermore, healthy controls (HCs) and MDD patients underwent magnetic resonance imaging (MRI) scans for functional connectivity (FC) analysis. After 6 weeks of treatment, the SSRIs + AIA group showed a greater reduction in HAMD-17 score than the SSRIs + SIA group (MD, -4.9 [CI, -7.6 to -2.2], P < 0.001) and SSRIs group (MD, -5.1 [CI, -7.8 to -2.3], P < 0.001). No serious adverse events occurred. SSRIs + AIA resulted in lower incidences of palpitations (vs.SSRIs + SIA: OR, 0.1% [CI, 0.0-1.0%]; vs. SSRIs: OR, 0.1% [CI, 0.0-0.7%]; P < 0.05), somnolence (vs.SSRIs + SIA: OR, 0.1% [CI, 0.0-0.9%]; vs.SSRIs: OR, 0.1% [CI, 0.0-0.7%]; P < 0.05), and nausea (vs.SSRIs + SIA: OR, 0.1% [CI, 0.0-1.0%]; vs. SSRIs: OR, 0.1% [CI, 0.0-0.9%]; P < 0.05). MDD patients showed abnormal FCs, and IA enhanced FCs between striatum and frontal_inf_tri, and striatum and cerebellum in the MRI study. Overall, IA as adjunctive therapy provides clinical efficacy and safety for MDD, and it may exert antidepressant effects by modulating striatal FCs.
Collapse
Affiliation(s)
- Xiaoting Wu
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Mingqi Tu
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Department of Acupuncture, The Affiliated Hangzhou First People's Hospital, Hangzhou, China
| | - Zelin Yu
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijian Cao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Siying Qu
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Nisang Chen
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Junyan Jin
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Sangsang Xiong
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiajia Yang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Shuangyi Pei
- Department of Psychiatry, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia Wang
- Department of Psychiatry, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Shi
- Department of Acupuncture, The Affiliated Hangzhou First People's Hospital, Hangzhou, China
| | - Lishu Gao
- Department of Psychiatry, The Affiliated Hangzhou First People's Hospital, Hangzhou, China
| | - Jian Xie
- Department of Psychiatry, The Affiliated Hangzhou First People's Hospital, Hangzhou, China
| | - Xinwei Li
- Department of Acupuncture, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jianqiao Fang
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Xiaomei Shao
- Department of Acupuncture, The Third Clinical Medical College, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
2
|
Grigoryan GA. From memory disorders to the development of depression: A system approach. Biosystems 2025; 251:105440. [PMID: 40049440 DOI: 10.1016/j.biosystems.2025.105440] [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: 12/12/2024] [Revised: 02/12/2025] [Accepted: 02/26/2025] [Indexed: 05/06/2025]
Abstract
In this review, a hypothesis explaining the origin and genesis of depression development from the perspective of a holistic functional system of behavioral control is proposed. The core of the functional system is the memory apparatus, on which all other key components of the behavioral control system (sensory information, motivation, reinforcement, and motor activity) are interlocked. In the organization of memory traces (engrams) there are two inputs, sensory and motivational, through which the stimulus-stimulus (S-S) and stimulus-motor (S-R) engrams are formed. These engrams are organized and actualized by means of forward and backward conditional connections between cortical representations of sensory information and motivational structures of the brain. Through feedback connections from reinforcing (emotional) input to the memory apparatus, the S-S and S-R engrams are consolidated or weakened depending on the strength of reward or negative events. Depression begins with a breakdown in memory mechanisms. These breakdowns are related to problems with the three mentioned memory inputs: sensory, motivational, and reinforcing (emotional). Disruptions in sensory and motivational input lead to an inability to form new memory engrams, their actualization and retrieval. This creates difficulty in solving current and past unresolved problems, eliciting more accumulation and increasing difficulties in their solving. Unresolved tasks lead to weakening of the reinforcing input, and further impairment of consolidation of the acting engrams. Another reason for the weakening of reinforcing input is excessive action of directly harmful events or constant chronic stress. The review presents the current literature and some data from our laboratory in favor of each memory input's contribution and their impact on the development of depression, when they are problematic.
Collapse
Affiliation(s)
- Grigory A Grigoryan
- Department of Conditioned Reflexes and Physiology of Emotions, Institute of Higher Nervous Activity and Neurophysiology RAS, 5a Butlerov str., Moscow, 117485, Russian Federation.
| |
Collapse
|
3
|
Teichman EM, Hu J, Lin HY, Fisher-Foye RL, Blando A, Hu X, Kaniskan HÜ, Montgomery SE, Cai M, Parise LF, Wang J, Russo SJ, Han MH, Jin J, Morel C. Design and validation of novel brain-penetrant HCN channel inhibitors to ameliorate social stress-induced susceptible phenotype. Mol Psychiatry 2025:10.1038/s41380-025-02972-8. [PMID: 40199995 DOI: 10.1038/s41380-025-02972-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 12/16/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Major Depressive Disorder (MDD) is a devastating, multifactorial disease with limited pharmacological treatment options. Patients with MDD exhibit alterations in their dopamine (DA) signaling pathways through the midbrain ventral tegmental area (VTA). A similar observation is also detected in preclinical models of stress - mice exhibit behavioral and physiological impairments following chronic social defeat stress (CSDS). Prior studies demonstrate that CSDS-susceptible mice have increased VTA DA neuronal excitability, in part driven by an upregulation in hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels. Inhibiting HCN channels with known inhibitors such as Cilobradine alleviates the negative behavioral effects of CSDS. Here, we aimed to identify Cilobradine analogs with improved neural tropism and inhibitory efficacy. Two compounds, MS7710 and MS7712, differing by their left-hand side moieties, have a similar, potent inhibitory effect on VTA DA Ih currents as compared to Cilobradine, and a greater inhibitory effect than Cilobradine on VTA DA firing rate. We demonstrate that MS7710 and MS7712 have superior brain/plasma concentration ratios as compared to Cilobradine. They were efficacious at inhibiting VTA DA neuron firing rate and bursting activity in CSDS-susceptible male mice at lower doses than Cilobradine, which was recapitulated in female CSDS-susceptible mice with MS7710. Finally, we define that a single intraperitoneal injection of MS7710 ameliorates CSDS-induced social interaction deficits and reward-associated cognitive inflexibility for at least two weeks in male and female mice. These findings yield a novel HCN channel inhibitor with improved neural tropism and stress-alleviating effects that could provide a basis for future antidepressant drug discovery.
Collapse
Affiliation(s)
- Emily M Teichman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jianping Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Hsiao-Yun Lin
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Rachel L Fisher-Foye
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Anthony Blando
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Xiaoping Hu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - H Ümit Kaniskan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sarah E Montgomery
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Modendo Inc., 3415 Colorado Ave, Boulder, Colorado, 80303, USA
| | - Min Cai
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lyonna F Parise
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jun Wang
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, New York, NY, USA
| | - Scott J Russo
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Brain-Body Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology; Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, China.
| | - Jian Jin
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Carole Morel
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Anderson KR, Rogu PJ, Palumbo TB, Miwa JM. Abnormal response to chronic social defeat stress and fear extinction in a mouse model of Lynx2-based cholinergic dysregulation. Front Neurosci 2025; 19:1466166. [PMID: 40236946 PMCID: PMC11998120 DOI: 10.3389/fnins.2025.1466166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/27/2025] [Indexed: 04/17/2025] Open
Abstract
Nicotinic receptor signaling is influential in modulating appropriate responses to salient stimuli within a complex environment. The cholinergic neurotransmitter system drives attention to salient stimuli such as stressors, and aids in orchestrating the proper neural and behavioral responses. Dysregulation of this system, however, has been implicated in altered anxiety regulation and mood disorders. Among the multiple layers of regulation are protein modulators such as Lynx2/Lypd1, which provides negative nicotinic acetylcholine receptor regulation within anxiety-related circuits, such as the amygdala and medial prefrontal cortex, among other brain regions. Mice null for Lynx2/Lypd1 (Lynx2 KO) show elevated basal anxiety-like behavior in tests such as elevated plus maze, light-dark box and social interaction assays. Here, we queried how a line predisposed to basal anxiety-like behavior would respond to specific stressors, using validated models of experiential-based affective disorders such as fear extinction, acute and chronic social defeat stress assays. We discovered that Lynx2 KO mice demonstrate an inability to extinguish learned fear during fear extinction tests even during milder stress conditions. In social defeat studies, contrary to our predictions, the Lynx2 KO mice switched from a socially avoidant phenotype (which could be considered susceptible) before defeat to a social approach/resilient phenotype after defeat. Consistent with reports of the inverse relationship between resilience and BDNF levels, we observed reduced BDNF levels in the VTA of Lynx2 KO mice. Furthermore, we provide evidence for the functional role of α7 nicotinic receptor subtypes by phenotypic rescue of fear extinction and social defeat phenotypes by MLA antagonism of α7 nicotinic acetylcholine receptors, or by crossing with α7 nicotinic acetylcholine receptor null mutant mice. A stable physical interaction between LYNX2 and α7 nAChRs was observed by co-immunoprecipitation of complexes from mouse amygdalae extracts. Together, these data indicate that responses to specific stressors can become aberrant when baseline genetic factors predispose animals to anxiety dysregulation. These studies underscore the critical nature of well-regulated nicotinic receptor function in the adaptive response to environmental stressors.
Collapse
Affiliation(s)
| | | | | | - Julie M. Miwa
- Department of Biological and Chemical Sciences, Bethlehem, PA, United States
| |
Collapse
|
5
|
Breit S, Denier N, Mertse N, Walther S, Soravia LM, Federspiel A, Wiest R, Bracht T. The neurobiology of motivational anhedonia in patients with depression. Brain Imaging Behav 2025:10.1007/s11682-025-00999-7. [PMID: 40163222 DOI: 10.1007/s11682-025-00999-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Anhedonia is a core feature of depression. It contains a consummatory and a motivational aspect. Whilst much neuroimaging research in patients with depression focused on the consummatory aspect of anhedonia, less is known about its motivational aspect. This study aimed to explore the neurobiology of networks related to motivational anhedonia. Thirty-eight patients with major depressive disorder (MDD) and 19 healthy controls underwent diffusion-weighted and resting state functional magnetic resonance imaging (rs-fMRI). For assessment of motivational anhedonia, we summed the values of the CORE non-interactiveness score, and the items 1 (hopelessness) and 7 (work and activities) of the Hamilton Depression Rating Scale. Whole-brain voxel-wise statistical analysis of fractional anisotropy (FA) data was performed using Tract-Based Spatial Statistics (TBSS). Additionally, we performed a whole-brain comparison of integrated local correlation of rs-fMRI signal (LCOR), to investigate regional functional differences between patients and healthy controls. Whole brain correlations between motivational anhedonia and measures of structural and functional connectivity (FA, and LCOR) were calculated. TBSS-analyses revealed reduced FA in the left superior longitudinal fasciculus (SLF) in patients with MDD. LCOR was reduced in patients with depression in an adjacent cluster localized in bilateral precunei. Within patients, there was a positive correlation between motivational anhedonia and LCOR in the precunei and a negative correlation in bilateral sensorimotor areas. FA-values did not show significant correlations. These findings suggest that motivational anhedonia in depression is linked to alterations of functional connectivity within bilateral precunei. Observed white matter microstructural alterations in the SLF do not show such an association.
Collapse
Affiliation(s)
- Sigrid Breit
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
| | - Niklaus Denier
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
| | - Nicolas Mertse
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
| | - Sebastian Walther
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
- Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health, University Hospital of Würzburg, Würzburg, Germany
| | - Leila M Soravia
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
| | - Andrea Federspiel
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
| | - Roland Wiest
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland
- Institute of Diagnostic and Interventional Neuroradiology, University of Bern, Bern, Switzerland
| | - Tobias Bracht
- University Hospital of Psychiatry and Psychotherapy, University of Bern, Bern, Switzerland.
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, University of Bern, Bern, Switzerland.
| |
Collapse
|
6
|
Morris LS, Costi S, Hameed S, Collins KA, Stern ER, Chowdhury A, Morel C, Salas R, Iosifescu DV, Han MH, Mathew SJ, Murrough JW. Effects of KCNQ potassium channel modulation on ventral tegmental area activity and connectivity in individuals with depression and anhedonia. Mol Psychiatry 2025:10.1038/s41380-025-02957-7. [PMID: 40133425 DOI: 10.1038/s41380-025-02957-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 02/13/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025]
Abstract
Up to half of individuals with depression do not respond to first-line treatments, possibly due to a lack of treatment interventions informed by neurobiology. A novel therapeutic approach for depression has recently emerged from translational work targeting aberrant activity of ventral tegmental area (VTA) dopamine neurons via modulation of the KCNQ voltage-gated potassium channels. In this study, individuals with major depressive disorder (MDD) with elevated anhedonia were randomized to five weeks of the KCNQ channel opener, ezogabine (up to 900 mg/day) or placebo. Participants completed functional MRI during a monetary anticipation task and resting-state at baseline and at end-of-treatment. The clinical results were reported previously. Here, we examined VTA activity during monetary anticipation and resting-state functional connectivity between the VTA and the ventromedial prefrontal cortex (mesocortical pathway) and ventral striatum (mesolimbic pathway) at baseline and end-of-treatment. Results indicated a significant drug-by-time interaction in VTA activation during anticipation (F(1,34) = 4.36, p = 0.044), where VTA activation was reduced from pre-to-post ezogabine, compared to placebo. Mesocortical functional connectivity was also higher in depressed participants at baseline compared to a healthy control group (t(56) = 2.68, p = 0.01) and associated with VTA hyper-activity during task-based functional MRI at baseline (R = 0.352, p = 0.033). Mesocortical connectivity was also reduced from pre-to-post ezogabine, compared to placebo (significant drug-by-time interaction, F(1,33) = 4.317, p = 0.046). Together this translational work is consistent with preclinical findings highlighting VTA hyper-activity in depression, and suggesting a mechanism of action for KCNQ channel openers in normalizing this hyper-activity in individuals with both depression and anhedonia.
Collapse
Affiliation(s)
- Laurel S Morris
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
| | - Sara Costi
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Psychiatry, University of Oxford, Oxford, UK
- Oxford Health NHS Foundation Trust, Warneford Hospital, Oxford, UK
| | - Sara Hameed
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Emily R Stern
- Nathan Kline Institute, Orangeburg, NY, USA
- New York University Grossman School of Medicine, New York, NY, USA
| | - Avijit Chowdhury
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Carole Morel
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ramiro Salas
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- The Menninger Clinic, Houston, TX, USA
- Neuroscience Department, Baylor College of Medicine, Houston, TX, USA
- Center for Translational Research on Inflammatory Diseases, Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - Dan V Iosifescu
- Nathan Kline Institute, Orangeburg, NY, USA
- New York University Grossman School of Medicine, New York, NY, USA
| | - Ming-Hu Han
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, PR China
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjay J Mathew
- Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
- The Menninger Clinic, Houston, TX, USA
- Michael E. Debakey VA Medical Center, Houston, TX, USA
| | - James W Murrough
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, USA.
- VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
7
|
Morris LS, Beltrán JM, Murrough JW, Morel C. Cross-species dissection of the modular role of the ventral tegmental area in depressive disorders. Neuroscience 2025; 569:248-266. [PMID: 39914519 PMCID: PMC11885014 DOI: 10.1016/j.neuroscience.2025.02.008] [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: 09/19/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Depressive disorders, including major depressive disorder (MDD), represent one of the most prevalent set of disorders worldwide. MDD is characterized by a range of cognitive, behavioral, and neurobiological changes that contribute to the vast array of symptom profiles that make this disorder particularly difficult to treat. A multitude of established evidence suggests a role for the dopamine system, stemming in part from the ventral tegmental area (VTA), in mediating symptoms and behavioral changes that underlie depression. Developments in cutting-edge technologies in pre-clinical models of depressive phenotypes, such as retrograde tracing, electrophysiological recordings, immunohistochemistry, and molecular profiling, have allowed a deeper characterization of singular VTA neuron molecular, physiological, and projection properties. These developments have highlighted that the VTA is not a homogenous cell population but instead comprises vast cellular diversity that underscores its modular role across various functions related to reward processing, aversion, salience processing, learning and motivation. In this review, we begin by introducing the various cell types and brain regions that comprise the VTA circuitry. Then, we introduce the role of the VTA in reward processing as it compares to aversion processing. Next, we characterize distinct neural pathways within the VTA circuitry to understand the effects of chronic social and non-social stress and tie together how these neurobiological changes manifest into specific behavioral phenotypes. Finally, we relate these preclinical findings to clinical findings to parse the heterogeneity of depressive phenotypes and explain the efficacy of recent novel pharmacological interventions that may target the VTA in MDD.
Collapse
Affiliation(s)
- L S Morris
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Nuffield Department of Clinical Neurosciences, University of Oxford, UK; Department of Experimental Psychology, University of Oxford, UK.
| | - J M Beltrán
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States
| | - J W Murrough
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai New York NY United States; Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States; VISN 2 Mental Illness Research, Education, and Clinical Center (MIRECC), James J. Peters VA Medical Center Bronx NY United States
| | - C Morel
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai New York NY United States.
| |
Collapse
|
8
|
Nakatsuka D, Suwa T, Deguchi Y, Fujita Y, Tashima R, Ohnami S, Kawashima H, Oishi N, Ogawa K, Yamakawa H, Murai T. Fine-tuning of dopamine receptor signaling with aripiprazole counteracts ketamine's dissociative action, but not its antidepressant effect. Transl Psychiatry 2025; 15:77. [PMID: 40057507 PMCID: PMC11890785 DOI: 10.1038/s41398-025-03284-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/18/2025] [Accepted: 02/11/2025] [Indexed: 05/13/2025] Open
Abstract
Ketamine, a rapid-acting antidepressant, has undesirable psychotomimetic effects, including a dissociative effect. There is currently no effective strategy to suppress these side effects while preserving its antidepressant effect. Here, we investigated the effects of a D2/D3 receptor antagonist and partial agonists on the psychotomimetic and antidepressant effects of ketamine in mice and humans. Aripiprazole, a partial agonist, attenuated the psychotomimetic effect, but maintaining and even enhancing the antidepressant-like effect of ketamine in the forced swim test, whereas raclopride, an antagonist, suppressed both effects in mice. Brain-wide Fos mapping and its network analysis suggested the ventral tegmental area (VTA) as a critical region for distinguishing the effects of aripiprazole and raclopride. In the chronic stress model, local infusion of raclopride into the VTA inhibited ketamine's antidepressant-like effect, accompanied by activation of dopaminergic neurons, suggesting the inhibitory effect of VTA activation on the antidepressant-like effect of ketamine. Consistently, systemic injections of raclopride and brexpiprazole, a partial agonist similar to aripiprazole but closer to an antagonist (lower Emax), activated dopaminergic neurons in the VTA and suppressed ketamine's antidepressant-like effect in the model when co-administered with ketamine, whereas aripiprazole didn't. In line with these results, in a single-arm, double-blinded clinical study of sequential treatments in depressed patients (N = 9), co-administration of 12 mg of aripiprazole suppressed the dissociative symptoms induced by ketamine while maintaining its antidepressant effects. Together, these findings suggest that fine-tuning dopamine receptor signaling with aripiprazole allows selective suppression of ketamine-induced dissociation preserving its antidepressant effects, and that the combined use of aripiprazole and ketamine may be a preferred therapy for treatment-resistant depression.
Collapse
Affiliation(s)
- Daiki Nakatsuka
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan
- Ping An-Shionogi Co., Ltd, Shanghai, China
| | - Taro Suwa
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuichi Deguchi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan
| | - Yoshihisa Fujita
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryoichi Tashima
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan
| | - Soichiro Ohnami
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan
| | - Hirotsugu Kawashima
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Naoya Oishi
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koichi Ogawa
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan
| | - Hidekuni Yamakawa
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Laboratory for Drug Discovery and Disease Research, SHIONOGI & CO., LTD, Osaka, Japan.
- Ping An-Shionogi Co., Ltd, Shanghai, China.
| | - Toshiya Murai
- SK Project, Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan.
- Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
9
|
Zhang L, Ji M, Sun Y, Wang Q, Jin M, Wang S, Sun H, Zhang H, Huang D. VTA dopaminergic neurons involved in chronic spared nerve injury pain-induced depressive-like behavior. Brain Res Bull 2025; 222:111261. [PMID: 39956400 DOI: 10.1016/j.brainresbull.2025.111261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Affective disorders, such as depression, are commonly associated with the development of chronic pain, but the underlying mechanisms still remain unclear. The dopaminergic system, located in the midbrain, is considered one of the regions where algesia and emotional processing overlap. This suggests a structural basis hypothesis for the comorbidity of chronic pain and depression, highlighting the interplay between nociceptive and affective processing. But there are more and more evidences show that somatic and head/facial pain involve different neuronal overlap. In previous study, the research show that VTA dopaminergic system involved in pIONT surgery induced depressive-like behaviors in mice. But there still no evidence shows if chronic somatic pain will induce depressive-like behaviors and which neuronal circle pathway is underly. In this study, we assessed depressive-like behaviors and performed artificial interference of VTA (ventral tegmental area) dopaminergic neurons in a mouse model of chronic peripheral neuropathic pain induced by the spared nerve injury (SNI) model. After a 4-week duration of hyperalgesia and allodynia resulting from SNI surgery, social withdraw and other depressive-like behaviors were observed in the SNI group. Furthermore, the dopaminergic cells' excitability in VTA were significantly increased in SNI mice. The excitability alteration was improved play a key role in the development and modulation of the chronic peripheral neuropathic pain-induced depressive-like behaviors. It has been shown pain and affections have structural and functional circuits to interact with each other, therefore the neuroplastic changes and functional role of VTA dopaminergic neurons within these circuits may serve as potential targets for understanding and therapeutically addressing the development of depressive-like symptoms accompanied by prolonged pain syndromes in humans.
Collapse
Affiliation(s)
- Ludi Zhang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, 050017, Shijiazhuang, Hebei, PR China; College of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China; Identification Center of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Menghan Ji
- College of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Yufei Sun
- College of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Qingwu Wang
- Identification Center of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Mingyang Jin
- Identification Center of Forensic Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, PR China
| | - Shuling Wang
- The First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, PR China
| | - Hui Sun
- Department of Physiology, Binzhou Medical University, Yantai 264003, PR China
| | - Hailin Zhang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, 050017, Shijiazhuang, Hebei, PR China
| | - Dongyang Huang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, 050017, Shijiazhuang, Hebei, PR China; Institute of Chinese Integrative Medicine, Hebei Medical University, Shijiazhuang 050000, PR China.
| |
Collapse
|
10
|
Chen HR, Wang T, Shao J, Zhu HM, Chi-Zhang, Zhao T, Xu LH, Wang M, Li JJ, Zhu QL, Qi XM, Xu DX, Wang B, Meng XH. Paternal fenvalerate exposure causes depressive-like behaviour by altering Grb10 gene DNA methylation in adolescent offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117994. [PMID: 40043501 DOI: 10.1016/j.ecoenv.2025.117994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/05/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025]
Abstract
Fenvalerate, a typical pyrethroid pesticide, is a neurological toxicant. This study aimed to evaluate the effect of paternal exposure to fenvalerate on depressive-like behaviours in adolescent offspring. Depression-like behavior was determined by Sucrose Preference Test (SPT), Tail Suspension Test (TST) and Forced Swimming Test (FST) in adolescent offspring. The level of dopamine was reduced in the midbrain of fenvalerate-exposed adolescent offspring. Tyrosine hydroxylase (Th), a rate limiting enzyme for dopamine synthesis, was significantly reduced in the midbrain of adolescent offspring exposed to fenvalerate. And Th was decreased in the midbrain and hindbrain of fetuses exposed to fenvalerate. Transcriptome analysis revealed growth factor receptor-bound protein 10 (Grb10) was decreased in the fetal hindbrain exposed to fenvalerate. Grb10 mRNA and protein were reduced in the fetal hindbrain exposed to fenvalerate. Interestingly, in vitro experiments, Th was reduced by si-Grb10. Conversely, Th was increased by oe-Grb10. Mechanistically, the 5mC content of Grb10 gene at one CpG fragment was reduced in the fetal hindbrain exposed to fenvalerate. And the 5mC content of Grb10 gene at eighteen CpG sites was decreased in paternal sperm exposed to fenvalerate. In summary, paternal fenvalerate exposure causes depressive-like behavior by altering DNA methylation of Grb10 gene in the sperm.
Collapse
Affiliation(s)
- Hui-Ru Chen
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Tao Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course / Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Jing Shao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Hui-Min Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Chi-Zhang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Ting Zhao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Li-Hua Xu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Min Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Jing-Jing Li
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Qi-Long Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xi-Meng Qi
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China
| | - De-Xiang Xu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course / Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Bo Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course / Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Xiu-Hong Meng
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, Anhui 230032, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Environment and Population Health Across the Life Course / Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
11
|
Pérez-Garci E, Pysanenko K, Rizzi G, Studer F, Ulrich D, Fritzius T, Früh S, Porcu A, Besseyrias V, Melichar A, Gassmann M, Barkat TR, Tureček R, Tan KR, Bettler B. Binding of HCN channels to GABA B receptors in dopamine neurons of the VTA limits synaptic inhibition and prevents the development of anxiety. Neurobiol Dis 2025; 206:106831. [PMID: 39914775 DOI: 10.1016/j.nbd.2025.106831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
During GABAergic synaptic transmission, G protein-coupled GABAB receptors (GBRs) activate K+ channels that prolong the duration of inhibitory postsynaptic potentials (IPSPs). We now show that KCTD16, an auxiliary GBR subunit, anchors hyperpolarization-activated cyclic nucleotide-gated (HCN) channels containing HCN2/HCN3 subunits to GBRs. In dopamine neurons of the VTA (DAVTA neurons), this interaction facilitates activation of HCN channels via hyperpolarization during IPSPs, counteracting the GBR-mediated late phase of these IPSPs. Consequently, disruption of the GBR/HCN complex in KCTD16-/- mice leads to prolonged optogenetic inhibition of DAVTA neuron firing. KCTD16-/- mice exhibit increased anxiety-like behavior in response to stress - a behavior replicated by CRISPR/Cas9-mediated KCTD16 ablation in DAVTA neurons or by intra-VTA infusion of an HCN antagonist in wild-type mice. Our findings support that the retention of HCN channels at GABAergic synapses by GBRs in DAVTA neurons provides a negative feedback mechanism that restricts IPSP duration and mitigates the development of anxiety.
Collapse
Affiliation(s)
- Enrique Pérez-Garci
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Kateryna Pysanenko
- Department of Auditory Neuroscience, Institute of Experimental Medicine, CAS, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Giorgio Rizzi
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | - Florian Studer
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Daniel Ulrich
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Thorsten Fritzius
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Simon Früh
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Alessandra Porcu
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Valérie Besseyrias
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Adolf Melichar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, CAS, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Martin Gassmann
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Tania Rinaldi Barkat
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland
| | - Rostislav Tureček
- Department of Auditory Neuroscience, Institute of Experimental Medicine, CAS, Videnska 1083, 14220 Prague 4, Czech Republic
| | - Kelly R Tan
- Biozentrum, University of Basel, Spitalstrasse 41, CH-4056 Basel, Switzerland
| | - Bernhard Bettler
- Department of Biomedicine, Pharmazentrum, University of Basel, Klingelbergstrasse 50, CH-4056 Basel, Switzerland.
| |
Collapse
|
12
|
Duan Z, Zhao W, Tong Y, Coenen VA, Döbrössy MD. Acute and chronic gene expression activation following medial forebrain bundle DBS and selective dopamine pathway stimulation. Sci Rep 2025; 15:7131. [PMID: 40021746 PMCID: PMC11871370 DOI: 10.1038/s41598-025-91994-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
Deep brain stimulation (DBS) of the medial forebrain bundle (mfb) demonstrated anti-depressant effects both clinically and experimentally. Modulation of mesocorticolimbic dopaminergic (DA) activity could contribute-in part-to the therapeutic effects. By comparing selective and pathway specific midbrain DA optogenetic stimulation with the global, non-pathway specific mfb-DBS, the study explored changes in gene-expression of key biomarkers associated with neurocircuitry of depression. Rats received either optogenetic DAergic or mfb-DBS, delivered as acute/single or chronic/repeated stimulation. Micro-dissected regions were prepared for in situ hybridization targeting biomarkers of GABAergic, glutamatergic, and dopaminergic systems. Mfb-DBS mediated DA independent pathway increased GABAergic biomarkers (GABAA, GAD1) in frontal and accumbal regions, not in midbrain. The combinations of low frequency/high pulse width and high frequency/low pulse width stimulation generally increased biomarker expression similarly, but chronic/repetitive stimulation had no accumulative effect. Interestingly, unilateral stimulation had bilateral effects, but stimulation modalities had little impact on DAT and Vglut2 expression. In conclusion, both low and high frequency, acute/single and chronic/repetitive mfb-DBS-but not selective optogenetic stimulation -activated gene expression of biomarkers associated with GABAergic transmission. The increased expression was transitory and less chronic than predicted. Importantly, the study provides evidence that the anti-depressant therapeutic effects of clinical medial forebrain bundle DBS occurs-in part-be via modulation of GABAergic signalling which in turn could regulate the release of dopamine in frontal and accumbal regions. In addition, clinical implication of the data is that unilateral stimulation had bilateral consequences on the gene expression, although the physiological and functional sequelae of this are yet unknown.
Collapse
Affiliation(s)
- Zhuo Duan
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wen Zhao
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yixin Tong
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Volker A Coenen
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Máté D Döbrössy
- Laboratory of Stereotaxy and Interventional Neurosciences, Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Breisacher Str. 64, 79106, Freiburg, Germany.
- Department of Stereotactic and Functional Neurosurgery, University Freiburg - Medical Centre, Freiburg, Germany.
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
13
|
Li S, Zhang J, Li J, Hu Y, Zhang M, Wang H. Optogenetics and chemogenetics: key tools for modulating neural circuits in rodent models of depression. Front Neural Circuits 2025; 19:1516839. [PMID: 40070557 PMCID: PMC11893610 DOI: 10.3389/fncir.2025.1516839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Optogenetics and chemogenetics are emerging neuromodulation techniques that have attracted significant attention in recent years. These techniques enable the precise control of specific neuronal types and neural circuits, allowing researchers to investigate the cellular mechanisms underlying depression. The advancement in these techniques has significantly contributed to the understanding of the neural circuits involved in depression; when combined with other emerging technologies, they provide novel therapeutic targets and diagnostic tools for the clinical treatment of depression. Additionally, these techniques have provided theoretical support for the development of novel antidepressants. This review primarily focuses on the application of optogenetics and chemogenetics in several brain regions closely associated with depressive-like behaviors in rodent models, such as the ventral tegmental area, nucleus accumbens, prefrontal cortex, hippocampus, dorsal raphe nucleus, and lateral habenula and discusses the potential and challenges of optogenetics and chemogenetics in future research. Furthermore, this review discusses the potential and challenges these techniques pose for future research and describes the current state of research on sonogenetics and odourgenetics developed based on optogenetics and chemogenetics. Specifically, this study aimed to provide reliable insights and directions for future research on the role of optogenetics and chemogenetics in the neural circuits of depressive rodent models.
Collapse
Affiliation(s)
- Shaowei Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianying Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiehui Li
- Shengli Oilfield Central Hospital, Dongying Rehabilitation Hospital, Dongying, China
| | - Yajie Hu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mingkuan Zhang
- College of Medical and Healthcare, Linyi Vocational College, Linyi, China
| | - Haijun Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
14
|
Lupinsky D, Nasseef MT, Parent C, Craig K, Diorio J, Zhang TY, Meaney MJ. Resting-state fMRI reveals altered functional connectivity associated with resilience and susceptibility to chronic social defeat stress in mouse brain. Mol Psychiatry 2025:10.1038/s41380-025-02897-2. [PMID: 39984680 DOI: 10.1038/s41380-025-02897-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 12/17/2024] [Accepted: 01/14/2025] [Indexed: 02/23/2025]
Abstract
Chronic stress is a causal antecedent condition for major depressive disorder and associates with altered patterns of neural connectivity. There are nevertheless important individual differences in susceptibility to chronic stress. How functional connectivity (FC) amongst interconnected, depression-related brain regions associates with resilience and susceptibility to chronic stress is largely unknown. We used resting-state functional magnetic resonance imaging (rs-fMRI) to examine FC between established depression-related regions in susceptible (SUS) and resilient (RES) adult mice following chronic social defeat stress (CSDS). Seed-seed FC analysis revealed that the ventral dentate gyrus (vDG) exhibited the greatest number of FC group differences with other stress-related limbic brain regions. SUS mice showed greater FC between the vDG and subcortical regions compared to both control (CON) or RES groups. Whole brain vDG seed-voxel analysis supported seed-seed findings in SUS mice but also indicated significantly decreased FC between the vDG and anterior cingulate area compared to CON mice. Interestingly, RES mice exhibited enhanced FC between the vDG and anterior cingulate area compared to SUS mice. Moreover, RES mice showed greater FC between the infralimbic prefrontal cortex and the nucleus accumbens shell compared to CON mice. These findings indicate unique differences in FC patterns in phenotypically distinct SUS and RES mice that could represent a neurobiological basis for depression, anxiety, and negative-coping behaviors that are associated with exposure to chronic stress.
Collapse
Affiliation(s)
- Derek Lupinsky
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
| | - Md Taufiq Nasseef
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
- Department of Mathematics, College of Science and Humanity Studies, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Carine Parent
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada
| | - Kelly Craig
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Josie Diorio
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Tie-Yuan Zhang
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada.
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, QC, Canada.
| | - Michael J Meaney
- Douglas Hospital Research Centre, Department of Psychiatry, McGill University, Montréal, QC, Canada.
- Translational Neuroscience Program, Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Brain-Body Initiative, Agency for Science, Technology & Research, Singapore, Singapore.
| |
Collapse
|
15
|
Krauklis SA, Towers AE, York JM, Baynard T, Gainey SJ, Freund GG, Steelman AJ. Mouse Testing Methods in Psychoneuroimmunology: Measuring Behavioral Responses. Methods Mol Biol 2025; 2868:163-203. [PMID: 39546231 DOI: 10.1007/978-1-0716-4200-9_10] [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: 11/17/2024]
Abstract
The field of psychoneuroimmunology (PNI) aims to uncover the processes and consequences of nervous, immune, and endocrine system relationships. Behavior is a consequence of such interactions and manifests from a complex interweave of factors including immune-to-neural and neural-to-immune communication. Often the signaling molecules involved during a particular episode of neuroimmune activation are not known, but behavioral response provides evidence that bioactives such as neurotransmitters and cytokines are perturbed. Immunobehavioral phenotyping is a first-line approach when examining the neuroimmune system and its reaction to immune stimulation or suppression. Behavioral response is significantly more sensitive than direct measurement of a single specific bioactive and can quickly and efficiently rule in or out relevance of a particular immune challenge or therapeutic to neuroimmunity. Classically, immunobehavioral research was focused on sickness symptoms related to bacterial infection, but neuroimmune activation is now a recognized complication of diseases and disorders ranging from cancer to diabesity to Alzheimer's. Immunobehaviors include lethargy, loss of appetite, and disinterest in social activity/surrounding environment. In addition, neuroimmune activation can diminish physical activity, precipitate feelings of depression and anxiety, and impair cognitive and executive function. Provided is a detailed overview of behavioral tests frequently used to examine neuroimmune activation in mice with a special emphasis on pre-experimental conditions that can confound or prevent successful immunobehavioral experimentation.
Collapse
Affiliation(s)
- Steven A Krauklis
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Albert E Towers
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jason M York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tracy Baynard
- Academic Affairs, University of Massachusetts-Boston, Boston, MA, USA
| | - Stephen J Gainey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Gregory G Freund
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| |
Collapse
|
16
|
Mancini C, Babicola L, Chila G, Di Segni M, Municchi D, D’Addario SL, Spoleti E, Passeri A, Cifani C, Andolina D, Cabib S, Ferlazzo F, Iosa M, Rossi R, Di Lorenzo G, Renzi M, Ventura R. Secure attachment to caregiver prevents adult depressive symptoms in a sex-dependent manner: A translational study. iScience 2024; 27:111328. [PMID: 39758994 PMCID: PMC11700650 DOI: 10.1016/j.isci.2024.111328] [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: 07/29/2024] [Revised: 09/13/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025] Open
Abstract
Although clinically relevant, evidence for a protective effect of early secure attachment against the development of depressive symptoms in adulthood is still inconsistent. This study used a translational approach to overcome this limitation. The analysis of a non-clinical adult population revealed a moderating effect of secure attachment on depressive symptoms in women only. Thus, we tested the causal link between early attachment with caregiver and adult depressive-like phenotypes in a mouse model of early adversities that is especially effective in females. Repeated cross fostering (RCF) in the first postnatal days prevented the development of pups' secure attachment with the caregiver as tested in a rodent version of the "strange situation"-the standard human test-induced depressive-like behaviors and altered activity of the ventral tegmental area dopamine neurons in adulthood. Finally, a stable alternative caregiver during the RCF experience prevented all these effects, modeling human "earned attachment."
Collapse
Affiliation(s)
- Camilla Mancini
- University of Camerino, School of Pharmacy, Pharmacology Unit, Camerino, Italy
| | | | - Gilda Chila
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Matteo Di Segni
- Child Psychopathology Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Diana Municchi
- Department of Psychology, Sapienza University, Rome, Italy
| | | | - Elena Spoleti
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Alice Passeri
- Department of Psychology, Sapienza University, Rome, Italy
| | - Carlo Cifani
- University of Camerino, School of Pharmacy, Pharmacology Unit, Camerino, Italy
| | - Diego Andolina
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Simona Cabib
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Fabio Ferlazzo
- Department of Psychology, Sapienza University, Rome, Italy
| | - Marco Iosa
- IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, Sapienza University, Rome, Italy
| | - Rodolfo Rossi
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Giorgio Di Lorenzo
- Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Massimiliano Renzi
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Rossella Ventura
- Department of Psychology, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| |
Collapse
|
17
|
Wang J, Su M, Zhang D, Zhang L, Niu C, Li C, You S, Sang Y, Zhang Y, Du X, Zhang H. The cation channel mechanisms of subthreshold inward depolarizing currents in the mice VTA dopaminergic neurons and their roles in the chronic-stress-induced depression-like behavior. eLife 2024; 12:RP88319. [PMID: 39642080 PMCID: PMC11623934 DOI: 10.7554/elife.88319] [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: 12/08/2024] Open
Abstract
The slow-intrinsic-pacemaker dopaminergic (DA) neurons originating in the ventral tegmental area (VTA) are implicated in various mood- and emotion-related disorders, such as anxiety, fear, stress and depression. Abnormal activity of projection-specific VTA DA neurons is the key factor in the development of these disorders. Here, we describe the crucial role of the NALCN and TRPC6, non-selective cation channels in mediating the subthreshold inward depolarizing current and driving the firing of action potentials of VTA DA neurons in physiological conditions. Furthermore, we demonstrate that down-regulation of TRPC6 protein expression in the VTA DA neurons likely contributes to the reduced activity of projection-specific VTA DA neurons in chronic mild unpredictable stress (CMUS) depressive mice. In consistent with these, selective knockdown of TRPC6 channels in the VTA DA neurons conferred mice with depression-like behavior. This current study suggests down-regulation of TRPC6 expression/function is involved in reduced VTA DA neuron firing and chronic stress-induced depression-like behavior of mice.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- Department of Chinese Medicinal Chemistry, Hebei University of Chinese MedicineShijiazhuangChina
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical UniversityShijiazhuangChina
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical UniversityShijiazhuangChina
| | - Min Su
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- Yiling Pharmaceutical CompanyShijiazhuangChina
| | - Dongmei Zhang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- Department of Clinical Pharmacy, Xingtai Ninth HospitalXingtaiChina
| | - Ludi Zhang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
| | - Chenxu Niu
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
| | - Chaoyi Li
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
| | - Shuangzhu You
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
| | - Yuqi Sang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- College of Chemical Engineering, Shijiazhuang UniversityShijiazhuangChina
- Shijiazhuang Key Laboratory of Targeted Drugs Research and Efficacy EvaluationShijiazhuangChina
| | - Yongxue Zhang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- Department of Pharmacy, Handan First HospitalHandanChina
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical UniversityShijiazhuangChina
- Department of Psychiatry, The First Hospital of Hebei Medical University, Mental Health Institute of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
18
|
Zhukovskaya A, Zimmerman CA, Willmore L, Pan-Vazquez A, Janarthanan SR, Lynch LA, Falkner AL, Witten IB. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. Neuron 2024; 112:3940-3956.e10. [PMID: 39393349 PMCID: PMC11624084 DOI: 10.1016/j.neuron.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 07/04/2024] [Accepted: 09/08/2024] [Indexed: 10/13/2024]
Abstract
Some individuals are susceptible to chronic stress, and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After (but not before) chronic social defeat stress, the LHb is active when susceptible mice are in proximity of the aggressor strain. During stress, activity is higher in susceptible mice during aggressor interactions, and activation biases mice toward susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical vs. cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
Affiliation(s)
- Anna Zhukovskaya
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | - Lindsay Willmore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | | | | | - Laura A Lynch
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Annegret L Falkner
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Ilana B Witten
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA; Howard Hughes Medical Institute, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
19
|
Ben Shaul T, Frenkel D, Gurevich T. The Interplay of Stress, Inflammation, and Metabolic Factors in the Course of Parkinson's Disease. Int J Mol Sci 2024; 25:12409. [PMID: 39596474 PMCID: PMC11594997 DOI: 10.3390/ijms252212409] [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: 09/29/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative condition for which there are symptomatic treatments but no disease-modifying therapies (DMTs). Extensive research over the years has highlighted the need for a multi-target DMT approach in PD that recognizes the various risk factors and their intricate interplay in contributing to PD-related neurodegeneration. Widespread risk factors, such as emotional stress and metabolic factors, have increasingly become focal points of exploration. Our review aims to summarize interactions between emotional stress and selected key players in metabolism, such as insulin, as potential mechanisms underlying neurodegeneration in PD.
Collapse
Affiliation(s)
- Tal Ben Shaul
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dan Frenkel
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tanya Gurevich
- Movement Disorders Center, Neurological Institute, Tel Aviv Medical Center, Tel Aviv 6423906, Israel;
- School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
20
|
Stanisavljević Ilić A, Filipović D. Mapping of c-Fos Expression in Rat Brain Sub/Regions Following Chronic Social Isolation: Effective Treatments of Olanzapine, Clozapine or Fluoxetine. Pharmaceuticals (Basel) 2024; 17:1527. [PMID: 39598437 PMCID: PMC11597560 DOI: 10.3390/ph17111527] [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: 10/15/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
The c-Fos as a marker of cell activation is used to identify brain regions involved in stimuli processing. This review summarizes a pattern of c-Fos immunoreactivity and the overlapping brain sub/regions which may provide hints for the identification of neural circuits that underlie depressive- and anxiety-like behaviors of adult male rats following three and six weeks of chronic social isolation (CSIS), relative to controls, as well as the antipsychotic-like effects of olanzapine (Olz), and clozapine (Clz), and the antidepressant-like effect of fluoxetine (Flx) in CSIS relative to CSIS alone. Additionally, drug-treated controls relative to control rats were also characterized. The overlapping rat brain sub/regions with increased expression of c-Fos immunoreactivity following three or six weeks of CSIS were the retrosplenial granular cortex, c subregion, retrosplenial dysgranular cortex, dorsal dentate gyrus, paraventricular nucleus of the thalamus (posterior part, PVP), lateral/basolateral (LA/BL) complex of the amygdala, caudate putamen, and nucleus accumbens shell. Increased activity of the nucleus accumbens core following exposure of CSIS rats either to Olz, Clz, and Flx treatments was found, whereas these treatments in controls activated the LA/BL complex of the amygdala and PVP. We also outline sub/regions that might represent potential neuroanatomical targets for the aforementioned antipsychotics or antidepressant treatments.
Collapse
Affiliation(s)
| | - Dragana Filipović
- Department of Molecular Biology and Endocrinology, “VINČA” Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
21
|
Díez-Solinska A, De Miguel Z, Azkona G, Vegas O. Behavioral coping with chronic defeat stress in mice: A systematic review of current protocols. Neurobiol Stress 2024; 33:100689. [PMID: 39628708 PMCID: PMC11612788 DOI: 10.1016/j.ynstr.2024.100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/18/2024] [Accepted: 11/06/2024] [Indexed: 12/06/2024] Open
Abstract
Social stress is the most significant source of chronic stress in humans and is commonly associated with health impairment. Individual differences in the behavioral coping responses to stress have been proposed to mediate the negative effects of stress on physical, behavioral and mental health. Animal models, particularly mice, offer valuable insights into the physiological and neurobiological correlates of behavioral coping strategies in response to chronic social stress. Here we aim to identify differences and similarities among stress protocols in mice, with particular attention to how neuroendocrine and/or behavioral responses vary according to different coping strategies, while highlighting the need for standardized approaches in future research. A systematic review was undertaken following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA statement). A total of 213 references were identified by electronic search, and after the screening, 18 articles were found to meet all the established criteria. We analyzed differences in the stress protocol, the characterization and classification of coping strategies and the physiological and behavioral differences according to coping. The results show that differences in behavioural expression under chronic social stress (coping) may also be associated with physiological differences and differential susceptibility to disease. However, this review also underlines the importance of a cautious interpretation of the results obtained. The lack of consistency in the nomenclature and procedures associated with the study of coping strategies for social stress, as well as the absence of a uniform classification, highlight the importance of using a common language when approaching the study of coping strategies. Thereby, this review encourages the development of a more defined method and criteria for assessing coping strategies, based on both behavioral and biological indicators.
Collapse
Affiliation(s)
- Alina Díez-Solinska
- Department of Basic Psychological Processes and Their Development, University of the Basque Country UPV/EHU, 20018, Donostia-San Sebastian, Spain
| | - Zurine De Miguel
- Department of Psychology, California State University, Monterey Bay, CA, USA
- Department of Health Sciences, Public University of Navarre UPNA, 31006, Pamplona, Spain
| | - Garikoitz Azkona
- Department of Basic Psychological Processes and Their Development, University of the Basque Country UPV/EHU, 20018, Donostia-San Sebastian, Spain
| | - Oscar Vegas
- Department of Basic Psychological Processes and Their Development, University of the Basque Country UPV/EHU, 20018, Donostia-San Sebastian, Spain
- Biogipuzkoa Health Research Institute, 20014, Donostia-San Sebastian, Spain
| |
Collapse
|
22
|
Zhukovskaya A, Christopher Z, Willmore L, Pan Vazquez A, Janarthanan S, Falkner A, Witten I. Heightened lateral habenula activity during stress produces brainwide and behavioral substrates of susceptibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565681. [PMID: 39005438 PMCID: PMC11244933 DOI: 10.1101/2023.11.06.565681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Some individuals are susceptible to the experience of chronic stress and others are more resilient. While many brain regions implicated in learning are dysregulated after stress, little is known about whether and how neural teaching signals during stress differ between susceptible and resilient individuals. Here, we seek to determine if activity in the lateral habenula (LHb), which encodes a negative teaching signal, differs between susceptible and resilient mice during stress to produce different outcomes. After, but not before, chronic social defeat stress (CSDS), the LHb is active when susceptible mice are in the proximity of the aggressor strain. During stress itself, LHb activity is higher in susceptible mice during aggressor proximity, and activation of the LHb during stress biases mice towards susceptibility. This manipulation generates a persistent and widespread increase in the balance of subcortical versus cortical activity in susceptible mice. Taken together, our results indicate that heightened activity in the LHb during stress produces lasting brainwide and behavioral substrates of susceptibility.
Collapse
|
23
|
Oliver D, Chesney E, Cullen AE, Davies C, Englund A, Gifford G, Kerins S, Lalousis PA, Logeswaran Y, Merritt K, Zahid U, Crossley NA, McCutcheon RA, McGuire P, Fusar-Poli P. Exploring causal mechanisms of psychosis risk. Neurosci Biobehav Rev 2024; 162:105699. [PMID: 38710421 PMCID: PMC11250118 DOI: 10.1016/j.neubiorev.2024.105699] [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: 11/01/2023] [Revised: 02/17/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
Robust epidemiological evidence of risk and protective factors for psychosis is essential to inform preventive interventions. Previous evidence syntheses have classified these risk and protective factors according to their strength of association with psychosis. In this critical review we appraise the distinct and overlapping mechanisms of 25 key environmental risk factors for psychosis, and link these to mechanistic pathways that may contribute to neurochemical alterations hypothesised to underlie psychotic symptoms. We then discuss the implications of our findings for future research, specifically considering interactions between factors, exploring universal and subgroup-specific factors, improving understanding of temporality and risk dynamics, standardising operationalisation and measurement of risk and protective factors, and developing preventive interventions targeting risk and protective factors.
Collapse
Affiliation(s)
- Dominic Oliver
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK; Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
| | - Edward Chesney
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - Alexis E Cullen
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Clinical Neuroscience, Karolinska Institutet, Sweden
| | - Cathy Davies
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Amir Englund
- Addictions Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 4 Windsor Walk, London SE5 8AF, UK
| | - George Gifford
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Sarah Kerins
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Paris Alexandros Lalousis
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany
| | - Yanakan Logeswaran
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Biostatistics & Health Informatics, King's College London, London, UK
| | - Kate Merritt
- Division of Psychiatry, Institute of Mental Health, UCL, London, UK
| | - Uzma Zahid
- Department of Psychology, King's College London, London, UK
| | - Nicolas A Crossley
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychiatry, School of Medicine, Pontificia Universidad Católica de Chile, Chile
| | - Robert A McCutcheon
- Department of Psychiatry, University of Oxford, Oxford, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Oxford Health NHS Foundation Trust, Oxford, UK
| | - Philip McGuire
- Department of Psychiatry, University of Oxford, Oxford, UK; NIHR Oxford Health Biomedical Research Centre, Oxford, UK; OPEN Early Detection Service, Oxford Health NHS Foundation Trust, Oxford, UK
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection (EPIC) Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Psychiatry and Psychotherapy, Ludwig-Maximilian-University Munich, Munich, Germany; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; OASIS Service, South London and Maudsley NHS Foundation Trust, London SE11 5DL, UK
| |
Collapse
|
24
|
Song RX, Miao HT, Jia SY, Li WG, Liu JZ, Zhang W, Xing BR, Zhao JY, Zhang LM, Li XM. Hemorrhagic Shock and Resuscitation Causes Excessive Dopaminergic Signaling in the mPFC and Cognitive Dysfunction. Mol Neurobiol 2024; 61:3899-3910. [PMID: 38041715 DOI: 10.1007/s12035-023-03804-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
Peri-operative hemorrhagic shock and resuscitation (HSR), a severe traumatic stress, is closely associated with post-operative anxiety, depression, and cognitive dysfunction, subsequently causing a serious burden on families and society. Following the co-release of corticotropin-releasing factor and catecholamine, traumatic stress activates dopaminergic neurons, increasing the addictive behavior and neurocognitive impairment risks. This study investigates the association between cognitive dysfunction and dopaminergic neurons in the mPFC under HSR conditions. This study established an HSR model by bleeding and re-transfusion in the mice. After HSR exposure, a dopamine D1 receptor antagonist, SKF-83566, was administered intraperitoneally for three consecutive days. Novel object recognition (NOR), conditioned fearing (FC), and conditioned place preference (CPP) were used to assess cognitive changes 16 days after HSR exposure. Local field potential (LFP) in the mPFC was also investigated during the novel object exploration. Compared with the mice exposed to sham, there was a significant decrease in the object recognition index, a reduction in context- and tone-related freezing time, an increase in CPP values, a downregulation of β-power but upregulation of γ-power in the mPFC in the mice exposed to HSR. Moreover, the mice exposed to HSR showed significantly upregulated TH-positive cell number, cleaved caspase-1- and TH-positive cells, and interleukin (IL)-1β/18 expression in the mPFC compared with sham; SKF-83566 could partially reverse these alternations. The HSR caused excessive dopaminergic signaling and cognitive dysfunction in the mPFC, a condition that might be ameliorated using a dopamine D1 receptor inhibitor.
Collapse
Affiliation(s)
- Rong-Xin Song
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Hui-Tao Miao
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Shi-Yan Jia
- Anesthesia and Trauma Research Unit, Department of Anesthesiology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Wen-Guang Li
- Graduate School, Hebei Medical University, Shijiazhuang, China
| | - Ji-Zhen Liu
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bao-Rui Xing
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Jian-Yong Zhao
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China
| | - Li-Min Zhang
- Hebei Province Key Laboratory of Integrated Traditional and Western Medicine in Neurological Rehabilitation, Cangzhou, China.
| | - Xiao-Ming Li
- Department of Orthopedics, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine (Cangzhou No. 2 Hospital), Cangzhou, China.
| |
Collapse
|
25
|
Han Y, Ai L, Song L, Zhou Y, Chen D, Sha S, Ji R, Li Q, Bu Q, Pan X, Zhai X, Cui M, Duan J, Yang J, Chaudhury D, Hu A, Liu H, Han MH, Cao JL, Zhang H. Midbrain glutamatergic circuit mechanism of resilience to socially transferred allodynia in male mice. Nat Commun 2024; 15:4947. [PMID: 38858350 PMCID: PMC11164890 DOI: 10.1038/s41467-024-49340-8] [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: 08/10/2023] [Accepted: 05/28/2024] [Indexed: 06/12/2024] Open
Abstract
The potential brain mechanism underlying resilience to socially transferred allodynia remains unknown. Here, we utilize a well-established socially transferred allodynia paradigm to segregate male mice into pain-susceptible and pain-resilient subgroups. Brain screening results show that ventral tegmental area glutamatergic neurons are selectively activated in pain-resilient mice as compared to control and pain-susceptible mice. Chemogenetic manipulations demonstrate that activation and inhibition of ventral tegmental area glutamatergic neurons bi-directionally regulate resilience to socially transferred allodynia. Moreover, ventral tegmental area glutamatergic neurons that project specifically to the nucleus accumbens shell and lateral habenula regulate the development and maintenance of the pain-resilient phenotype, respectively. Together, we establish an approach to explore individual variations in pain response and identify ventral tegmental area glutamatergic neurons and related downstream circuits as critical targets for resilience to socially transferred allodynia and the development of conceptually innovative analgesics.
Collapse
Affiliation(s)
- Yi Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Lin Ai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Lingzhen Song
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Yu Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Dandan Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Sha Sha
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Ran Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Qize Li
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Qingyang Bu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Xiangyu Pan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Xiaojing Zhai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Mengqiao Cui
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Jiawen Duan
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China
| | - Junxia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - Dipesh Chaudhury
- Division of Science, New York University Abu Dhabi (NYUAD), Saadiyat Island, 129188, United Arab Emirates
| | - Ankang Hu
- The Animal Facility of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China
| | - He Liu
- Department of Anesthesiology, Huzhou Central Hospital, Huzhou, Zhejiang, 313000, PR China
| | - Ming-Hu Han
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China.
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, PR China.
| |
Collapse
|
26
|
Ren L, Yu J, Chen H, Luo J, Lv F, Min S. Alteration of hyperpolarization-activated cation current-mediated metaplasticity contributes to electroconvulsive shock-induced learning and memory impairment in depressed rats. Front Psychiatry 2024; 15:1365119. [PMID: 38911706 PMCID: PMC11190359 DOI: 10.3389/fpsyt.2024.1365119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Accompanied by a rapid and effective antidepressant effect, electroconvulsive shock (ECS) can also induce learning and memory impairment. Our previous research reported that metaplasticity is involved in this process. However, the mechanisms still remain unclear. This study investigated the role of I h current in the metaplastic changes and learning and memory impairment induced by ECS in depressive rats. METHODS Depressive rats received ECS after modelling using chronic unpredictable. ZD7288, a type of I h current inhibitor was used to verify the effect of I h current. The sucrose preference test and Morris water maze were used for behavior testing. Changes in metaplasticity was assessed with the LTD/LTP threshold by stimulation at different frequencies. Spontaneous and evoked action potentials (APs) were measured to confirm difference of neuronal excitability. Additionally, the amplitude of I h current was analyzed. RESULTS ECS exerts antidepressant effect, but also induce spatial learning and memory dysfunction. ECS up-regulates the LTD/LTP threshold. In rats treated with ECS, the frequency of spontaneous and evoked APs is significantly reduced. In addition, ECS induces changes in the intrinsic properties of AP, including a decrease of AP-half width and peak amplitude, and an increase in AP time to peak and post-hyperpolarization potential amplitude. In particular, ECS increases both instantaneous and steady-state I h currents. However, Inhibition of I h current with ZD7288 results in a relief of learning and memory impairment and a decrease in threshold, as well as a significant reversal of whole-cell electrophysiological changes. CONCLUSION ECS-induced learning and memory impairment is caused by neuronal hypoexcitability mediated metaplasticity, and upregulation of LTD/LTP threshold by an increase in I h current.
Collapse
Affiliation(s)
- Li Ren
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Yu
- Department of Psychiatry, Shanghai 10th People’s Hospital, Anesthesia and Brain Research Institute, Tongji University, Shanghai, China
| | - Hengsheng Chen
- Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jie Luo
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Su Min
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
27
|
Rappeneau V, Castillo Díaz F. Convergence of oxytocin and dopamine signalling in neuronal circuits: Insights into the neurobiology of social interactions across species. Neurosci Biobehav Rev 2024; 161:105675. [PMID: 38608828 DOI: 10.1016/j.neubiorev.2024.105675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 04/14/2024]
Abstract
Social behaviour is essential for animal survival, and the hypothalamic neuropeptide oxytocin (OXT) critically impacts bonding, parenting, and decision-making. Dopamine (DA), is released by ventral tegmental area (VTA) dopaminergic neurons, regulating social cues in the mesolimbic system. Despite extensive exploration of OXT and DA roles in social behaviour independently, limited studies investigate their interplay. This narrative review integrates insights from human and animal studies, particularly rodents, emphasising recent research on pharmacological manipulations of OXT or DA systems in social behaviour. Additionally, we review studies correlating social behaviour with blood/cerebral OXT and DA levels. Behavioural facets include sociability, cooperation, pair bonding and parental care. In addition, we provide insights into OXT-DA interplay in animal models of social stress, autism, and schizophrenia. Emphasis is placed on the complex relationship between the OXT and DA systems and their collective influence on social behaviour across physiological and pathological conditions. Understanding OXT and DA imbalance is fundamental for unravelling the neurobiological underpinnings of social interaction and reward processing deficits observed in psychiatric conditions.
Collapse
Affiliation(s)
- Virginie Rappeneau
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany.
| | - Fernando Castillo Díaz
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Universitaetsstr. 31, Regensburg 93053, Germany
| |
Collapse
|
28
|
Smith A, Rodrigues T, Wallace C, Mezher K, MacAulay B, Prowse R, Hyland L, Abizaid A. Growth Hormone Secretagogue Receptor (GHSR) Signaling in the Ventral Tegmental Area (VTA) Mediates Feeding Produced by Chronic Social Defeat Stress in Male Mice. Neuroscience 2024; 547:17-27. [PMID: 38583506 DOI: 10.1016/j.neuroscience.2024.03.022] [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: 12/20/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 04/09/2024]
Abstract
Ghrelin, a hormone secreted by the stomach, binds to the growth hormone secretagogue receptor (GHSR) in various brain regions to produce a number of behavioral effects that include increased feeding motivation. During social defeat stress, ghrelin levels rise in correlation with increased feeding and potentially play a role in attenuating the anxiogenic effects of social defeat. One region implicated in the feeding effects of ghrelin is the ventral tegmental area (VTA), a region implicated in reward seeking behaviors, and linked to social defeat in mice. Here we examined the role of GHSR signaling in the VTA in feeding behavior in mice exposed to social defeat stress. Male C57BL/J6 mice that were socially defeated once daily for 3 weeks ate more, had higher plasma ghrelin level and increased GHSR expression in the VTA compared to non-stressed mice. Socially defeated GHSR KO mice failed to increase their caloric intake in response to this stressor but rescue of GHSR expression in the VTA restored feeding responses. Finally, we pharmacologically blocked VTA GHSR signalling with JMV2959 infused via an indwelling VTA cannula connected to a minipump. Vehicle-treated mice increased their caloric intake during social defeat, but JMV2959-infusions attenuated feeding responses and increased anxiety-like behaviors. The data suggest that GHSR signalling in the VTA is critical for the increases in appetite observed during chronic social defeat stress. Furthermore, these data support the idea that GHSR signaling in the VTA may also have anxiolytic effects, and blocking GHSR in this region may result in an anxiety-like phenotype.
Collapse
Affiliation(s)
- Andrea Smith
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Trevor Rodrigues
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Caroline Wallace
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Karen Mezher
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Brenna MacAulay
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Rebecca Prowse
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Lindsay Hyland
- Carleton University, Neuroscience Department, Ottawa, ON, Canada
| | - Alfonso Abizaid
- Carleton University, Neuroscience Department, Ottawa, ON, Canada.
| |
Collapse
|
29
|
Spreen A, Alkhoury D, Walter H, Müller S. Optogenetic behavioral studies in depression research: A systematic review. iScience 2024; 27:109776. [PMID: 38726370 PMCID: PMC11079475 DOI: 10.1016/j.isci.2024.109776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/21/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Optogenetics has made substantial contributions to our understanding of the mechanistic underpinnings of depression. This systematic review employs quantitative analysis to investigate the impact of optogenetic stimulation in mice and rats on behavioral alterations in social interaction, sucrose consumption, and mobility. The review analyses optogenetic behavioral studies using standardized behavioral tests to detect behavioral changes induced via optogenetic stimulation in stressed or stress-naive mice and rats. Behavioral changes were evaluated as either positive, negative, or not effective. The analysis comprises the outcomes of 248 behavioral tests of 168 studies described in 37 articles, including negative and null results. Test outcomes were compared for each behavior, depending on the animal cohort, applied type of stimulation and the stimulated neuronal circuit and cell type. The presented synthesis contributes toward a comprehensive picture of optogenetic behavioral research in the context of depression.
Collapse
Affiliation(s)
- Anika Spreen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
- Experimental Biophysics, Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Henrik Walter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Sabine Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| |
Collapse
|
30
|
Montgomery SE, Li L, Russo SJ, Calipari ES, Nestler EJ, Morel C, Han MH. Mesolimbic Neural Response Dynamics Predict Future Individual Alcohol Drinking in Mice. Biol Psychiatry 2024; 95:951-962. [PMID: 38061466 DOI: 10.1016/j.biopsych.2023.11.019] [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: 03/22/2023] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Individual variability in response to rewarding stimuli is a striking but understudied phenomenon. The mesolimbic dopamine system is critical in encoding the reinforcing properties of both natural reward and alcohol; however, how innate or baseline differences in the response dynamics of this circuit define individual behavior and shape future vulnerability to alcohol remain unknown. METHODS Using naturalistic behavioral assays, a voluntary alcohol drinking paradigm, in vivo fiber photometry, in vivo electrophysiology, and chemogenetics, we investigated how differences in mesolimbic neural circuit activity contribute to the individual variability seen in reward processing and, by proxy, alcohol drinking. RESULTS We first characterized heterogeneous behavioral and neural responses to natural reward and defined how these baseline responses predicted future individual alcohol-drinking phenotypes in male mice. We then determined spontaneous ventral tegmental area dopamine neuron firing profiles associated with responses to natural reward that predicted alcohol drinking. Using a dual chemogenetic approach, we mimicked specific mesolimbic dopamine neuron firing activity before or during voluntary alcohol drinking to link unique neurophysiological profiles to individual phenotype. We show that hyperdopaminergic individuals exhibit a lower neuronal response to both natural reward and alcohol that predicts lower levels of alcohol consumption in the future. CONCLUSIONS These findings reveal unique, circuit-specific neural signatures that predict future individual vulnerability or resistance to alcohol and expand the current knowledge base on how some individuals are able to titrate their alcohol consumption whereas others go on to engage in unhealthy alcohol-drinking behaviors.
Collapse
Affiliation(s)
- Sarah E Montgomery
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Long Li
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Erin S Calipari
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Departments of Pharmacology, Molecular Physiology and Biophysics, and Psychiatry and Behavioral Sciences, Vanderbilt Center for Addiction Research, Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Ming-Hu Han
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute and the Center for Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China.
| |
Collapse
|
31
|
Cuttoli RDD, Sweis BM. Ketamine reverses stress-induced hypersensitivity to sunk costs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593597. [PMID: 38798536 PMCID: PMC11118454 DOI: 10.1101/2024.05.12.593597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
How mood interacts with information processing in the brain is thought to mediate the maladaptive behaviors observed in depressed individuals. However, the neural mechanisms underlying impairments in emotion-cognition interactions are poorly understood. This includes influencing the balance between how past-sensitive vs. future-looking one is during decision-making. Recent insights from the field of neuroeconomics offer novel approaches to study changes in such valuation processes in a manner that is biologically tractable and readily translatable across species. We recently discovered that rodents are sensitive to "sunk costs" - a feature of higher cognition previously thought to be unique to humans. The sunk costs bias describes the phenomenon in which an individual overvalues and escalates commitment to continuing an ongoing endeavor, even if suboptimal, as a function of irrecoverable past (sunk) losses - information that, according to classic economic theory, should be ignored. In the present study, mice were exposed to chronic social defeat stress paradigm, a well-established animal model used for the study of depression. Mice were then tested on our longitudinal neuroeconomic foraging task, Restaurant Row. We found mice exposed to this severe stressor displayed an increased sensitivity to sunk costs, without altering overall willingness to wait. Mice were then randomly assigned to receive a single intraperitoneal injection of either saline or ketamine (20 mg/kg). We discovered that stress-induced hypersensitivity to sunk costs was renormalized following a single dose of ketamine. Interestingly, in non-defeated mice, ketamine treatment completely abolished sunk cost sensitivity, causing mice to no longer value irrecoverable losses during re-evaluation decisions who instead based choices solely on the future investment required to obtain a goal. These findings suggest that the antidepressant effects of ketamine may be mediated in part through changes in the processing of past-sensitive information during on-going decision-making, reducing its weight as a potential source of cognitive dissonance that could modulate behavior and instead promoting more future-thinking behavior.
Collapse
|
32
|
Hou G, Hao M, Duan J, Han MH. The Formation and Function of the VTA Dopamine System. Int J Mol Sci 2024; 25:3875. [PMID: 38612683 PMCID: PMC11011984 DOI: 10.3390/ijms25073875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/12/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The midbrain dopamine system is a sophisticated hub that integrates diverse inputs to control multiple physiological functions, including locomotion, motivation, cognition, reward, as well as maternal and reproductive behaviors. Dopamine is a neurotransmitter that binds to G-protein-coupled receptors. Dopamine also works together with other neurotransmitters and various neuropeptides to maintain the balance of synaptic functions. The dysfunction of the dopamine system leads to several conditions, including Parkinson's disease, Huntington's disease, major depression, schizophrenia, and drug addiction. The ventral tegmental area (VTA) has been identified as an important relay nucleus that modulates homeostatic plasticity in the midbrain dopamine system. Due to the complexity of synaptic transmissions and input-output connections in the VTA, the structure and function of this crucial brain region are still not fully understood. In this review article, we mainly focus on the cell types, neurotransmitters, neuropeptides, ion channels, receptors, and neural circuits of the VTA dopamine system, with the hope of obtaining new insight into the formation and function of this vital brain region.
Collapse
Affiliation(s)
- Guoqiang Hou
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mei Hao
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jiawen Duan
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ming-Hu Han
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China (M.H.); (J.D.)
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
33
|
Sang Y, Niu C, Xu J, Zhu T, You S, Wang J, Zhang L, Du X, Zhang H. PI4KIIIβ-Mediated Phosphoinositides Metabolism Regulates Function of the VTA Dopaminergic Neurons and Depression-Like Behavior. J Neurosci 2024; 44:e0555232024. [PMID: 38267258 PMCID: PMC10941068 DOI: 10.1523/jneurosci.0555-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/18/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024] Open
Abstract
Phosphoinositides, including phosphatidylinositol-4,5-bisphosphate (PIP2), play a crucial role in controlling key cellular functions such as membrane and vesicle trafficking, ion channel, and transporter activity. Phosphatidylinositol 4-kinases (PI4K) are essential enzymes in regulating the turnover of phosphoinositides. However, the functional role of PI4Ks and mediated phosphoinositide metabolism in the central nervous system has not been fully revealed. In this study, we demonstrated that PI4KIIIβ, one of the four members of PI4Ks, is an important regulator of VTA dopaminergic neuronal activity and related depression-like behavior of mice by controlling phosphoinositide turnover. Our findings provide new insights into possible mechanisms and potential drug targets for neuropsychiatric diseases, including depression. Both sexes were studied in basic behavior tests, but only male mice could be used in the social defeat depression model.
Collapse
Affiliation(s)
- Yuqi Sang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Chenxu Niu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jiaxi Xu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shanxi 710061, China
| | - Tiantian Zhu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Shuangzhu You
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Jing Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Ludi Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Collaborative Innovation Center of Hebei Province for Mechanism, Diagnosis and Treatment of Neuropsychiatric Diseases, Hebei Medical University, Shijiazhuang, Hebei 050011, China
- Department of Psychiatry, The First Hospital of Hebei Medical University, Mental Health Institute of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
34
|
Kc E, Islam J, Lee G, Park YS. Optogenetic Approach in Trigeminal Neuralgia and Potential Concerns: Preclinical Insights. Mol Neurobiol 2024; 61:1769-1780. [PMID: 37775720 DOI: 10.1007/s12035-023-03652-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/11/2023] [Indexed: 10/01/2023]
Abstract
The integration of optogenetics in the trigeminal pain circuitry broadens and reinforces existing pain investigations. Similar to research on spinal neuropathic pain, the exploration of the underlying determinants of orofacial pain is expanding. Optogenetics facilitates more direct, specific, and subtle investigations of the neuronal circuits involved in orofacial pain. One of the most significant concerns of both dentistry and medicine is trigeminal neuralgia (TN) management due to its substantial impact on a patient's quality of life. Our objective is to gather insights from preclinical studies conducted in TN employing an optogenetic paradigm, thereby extending the prospects for in-depth neurobiological research. This review highlights optogenetic research in trigeminal pain circuitry involving TN. We outline the central and peripheral regions associated with pain-that have been investigated using optogenetics in the trigeminal pain circuitry. The study further reports its scope and limitations as well as its potential for future applications from bench to bedside.
Collapse
Affiliation(s)
- Elina Kc
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Jaisan Islam
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Gabsang Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Young Seok Park
- Program in Neuroscience, Department of Medicine, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea.
- Department of Neurosurgery, Chungbuk National University Hospital, Cheongju, Republic of Korea.
| |
Collapse
|
35
|
Zhang L, Wang Y, Li S, Otani S, Chen F. Post-stress Social Interaction and 3-Cyano-N-(1,3-Diphenyl-1H-Pyrazol-5-yl) Benzamide Treatment Attenuate Depressive-like Behavior Induced by Repeated Social Defeat Stress. Neuroscience 2024; 538:11-21. [PMID: 38103860 DOI: 10.1016/j.neuroscience.2023.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Persistent stress increases the probability for developing depression significantly thereafter. Repeated social defeat stress is a widely used model to investigate depressive-like behavior in preclinical models. Hence, the repeated social defeat stress model provided an ideal animal model, through which the hypotheses of prevention and treatment can be investigated. We have successfully induced depressive-like behavior for male C57BL/6J mice with this model. Here, we reported that certain level of during-stress social interactions with single female or multiple male peer(s) exerted a positive role in preventing the development of depressive-like behavior induced by repeated social defeat stress. Our data suggested that the stress-susceptible mice may benefit from positive social interaction, which reduces the chance for depressive-like behavior development. Since numerous studies indicate that the metabotropic glutamate receptor 5 (mGluR5) plays an important role in various cognitive functions, we further investigate the treatment effect of 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) on the depressive-like behavior induced by repeated social defeat stress. Most importantly, robust anti-depressant effects have been achieved through modulating the mGluR5 function. We found that single oral dose administration of CDPPB (20 mg/kg), to some extent, alleviated the social avoidance behaviors for the stress-susceptible mice. Our data implies that the CDPPB, a positive allosteric modulator of mGluR5, is a promising anti-depressant candidate with limited side effect.
Collapse
Affiliation(s)
- Liangui Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Ying Wang
- Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Shengtian Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Satoru Otani
- Vision Institute, CNRS - INSERM - Sorbonne University, Paris 75012, France.
| | - Fujun Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
36
|
Chen Z, Ou Y, Liu F, Li H, Li P, Xie G, Cui X, Guo W. Increased brain nucleus accumbens functional connectivity in melancholic depression. Neuropharmacology 2024; 243:109798. [PMID: 37995807 DOI: 10.1016/j.neuropharm.2023.109798] [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: 09/23/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
BACKGROUND Melancholic depression, marked by typical symptoms of anhedonia, is regarded as a homogeneous subtype of major depressive disorder (MDD). However, little attention was paid to underlying mechanisms of melancholic depression. This study aims to examine functional connectivity of the reward circuit associated with anhedonia symptoms in melancholic depression. METHODS Fifty-nine patients with first-episode drug- naive MDD, including 31 melancholic patients and 28 non-melancholic patients, were recruited and underwent resting-state functional magnetic resonance imaging (rs-fMRI). Thirty-two healthy volunteers were recruited as controls. Bilateral nucleus accumbens (NAc) were selected as seed points to form functional NAc network. Then support vector machine (SVM) was used to distinguish melancholic patients from non-melancholic patients. RESULTS Relative to non-melancholic patients, melancholic patients displayed increased functional connectivity (FC) between bilateral NAc and right middle frontal gyrus (MFG) and between right NAc and left cerebellum lobule VIII. Compared to healthy controls, melancholic patients showed increased FC between right NAc and right lingual gyrus and between left NAc and left postcentral gyrus; non-melancholic patients had increased FC between bilateral NAc and right lingual gyrus. No significant correlations were observed between altered FC and clinical variables in melancholic patients. SVM results showed that FC between left NAc and right MFG could accurately distinguish melancholic patients from non-melancholic patients. CONCLUSION Melancholic depression exhibited different patterns of functional connectivity of the reward circuit relative to non-melancholic patients. This study highlights the significance of the reward circuit in the neuropathology of melancholic depression.
Collapse
Affiliation(s)
- Zhaobin Chen
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Yangpan Ou
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Feng Liu
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin 300000, China
| | - Huabing Li
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Ping Li
- Department of Psychiatry, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, China
| | - Guangrong Xie
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| | - Xilong Cui
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
37
|
Viudez-Martínez A, Torregrosa AB, Navarrete F, García-Gutiérrez MS. Understanding the Biological Relationship between Migraine and Depression. Biomolecules 2024; 14:163. [PMID: 38397400 PMCID: PMC10886628 DOI: 10.3390/biom14020163] [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: 12/27/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Migraine is a highly prevalent neurological disorder. Among the risk factors identified, psychiatric comorbidities, such as depression, seem to play an important role in its onset and clinical course. Patients with migraine are 2.5 times more likely to develop a depressive disorder; this risk becomes even higher in patients suffering from chronic migraine or migraine with aura. This relationship is bidirectional, since depression also predicts an earlier/worse onset of migraine, increasing the risk of migraine chronicity and, consequently, requiring a higher healthcare expenditure compared to migraine alone. All these data suggest that migraine and depression may share overlapping biological mechanisms. Herein, this review explores this topic in further detail: firstly, by introducing the common epidemiological and risk factors for this comorbidity; secondly, by focusing on providing the cumulative evidence of common biological aspects, with a particular emphasis on the serotoninergic system, neuropeptides such as calcitonin-gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), substance P, neuropeptide Y and orexins, sexual hormones, and the immune system; lastly, by remarking on the future challenges required to elucidate the etiopathological mechanisms of migraine and depression and providing updated information regarding new key targets for the pharmacological treatment of these clinical entities.
Collapse
Affiliation(s)
- Adrián Viudez-Martínez
- Hospital Pharmacy Service, Hospital General Dr. Balmis de Alicante, 03010 Alicante, Spain;
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández, 03550 San Juan de Alicante, Spain; (A.B.T.); (F.N.)
- Research Network on Primary Addictions, Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
38
|
Zhai X, Ai L, Chen D, Zhou D, Han Y, Ji R, Hu M, Wang Q, Zhang M, Wang Y, Zhang C, Yang JX, Hu A, Liu H, Cao JL, Zhang H. Multiple integrated social stress induces depressive-like behavioral and neural adaptations in female C57BL/6J mice. Neurobiol Dis 2024; 190:106374. [PMID: 38097092 DOI: 10.1016/j.nbd.2023.106374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/25/2023] [Accepted: 12/07/2023] [Indexed: 12/21/2023] Open
Abstract
Despite women representing most of those affected by major depression, preclinical studies have focused almost exclusively on male subjects, partially due to a lack of ideal animal paradigms. As the persistent need regarding the sex balance of neuroscience research and female-specific pathology of mental disorders surges, the establishment of natural etiology-based and systematically validated animal paradigms for depression with female subjects becomes an urgent scientific problem. This study aims to establish, characterize, and validate a "Multiple Integrated Social Stress (MISS)" model of depression in female C57BL/6J mice by manipulating and integrating daily social stressors that females are experiencing. Female C57BL/6J mice randomly experienced social competition failure in tube test, modified vicarious social defeat stress, unescapable overcrowding stress followed by social isolation on each day, for ten consecutive days. Compared with their controls, female MISS mice exhibited a relatively decreased preference for social interaction and sucrose, along with increased immobility in the tail suspension test, which could last for at least one month. These MISS mice also exhibited increased levels of blood serum corticosterone, interleukin-6 L and 1β. In the pharmacological experiment, MISS-induced dysfunctions in social interaction, sucrose preference, and tail suspension tests were amended by systematically administrating a single dose of sub-anesthetic ketamine, a rapid-onset antidepressant. Compared with controls, MISS females exhibited decreased c-Fos activation in their anterior cingulate cortex, prefrontal cortex, nucleus accumbens and some other depression-related brain regions. Furthermore, 24 h after the last exposure to the paradigm, MISS mice demonstrated a decreased center zone time in the open field test and decreased open arm time in the elevated plus-maze test, indicating anxiety-like behavioral phenotypes. Interestingly, MISS mice developed an excessive nesting ability, suggesting a likely behavioral phenotype of obsessive-compulsive disorder. These data showed that the MISS paradigm was sufficient to generate pathological profiles in female mice to mimic core symptoms, serum biochemistry and neural adaptations of depression in clinical patients. The present study offers a multiple integrated natural etiology-based animal model tool for studying female stress susceptibility.
Collapse
Affiliation(s)
- Xiaojing Zhai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Lin Ai
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Dandan Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Dongyu Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yi Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ran Ji
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Mengfan Hu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Qing Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Moruo Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yuxin Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Chunyan Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Ankang Hu
- Laboratory Animal Center of Xuzhou Medical University, Xuzhou Medical University, Xuzhou 221004, PR China
| | - He Liu
- Department of Anesthesiology & Clinical Research Center for Anesthesia and Perioperative Medicine, Huzhou Central Hospital, Huzhou 313003, China; The Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 313003, China; The Fifth School of Clinical Medicine, Zhejiang Chinese Medical University, Huzhou 313003, China; The Affiliated Central Hospital, Huzhou University, Huzhou 313003, China.
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Department of Anesthesiology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Hongxing Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China; NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
39
|
Yang Y, Wei X, Tian J, Zhu Y, Jia S, Shu Q. Scalp electroacupuncture targeting the VTA DA neurons to relieve negative emotions and promote the alleviation of chronic pain. Front Neurosci 2023; 17:1323727. [PMID: 38188034 PMCID: PMC10771389 DOI: 10.3389/fnins.2023.1323727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Object Chronic pain and negative emotions are often linked, and both can impact the reward circuit. The use of electroacupuncture (EA) has been found to regulate and improve these conditions. This study explores the potential mechanism of chronic pain relief by adding acupoints with emotional regulation effect to the basis of routine EA analgesia, to optimize the acupoint compatibility scheme of EA in the treatment of analgesia. Method For this study, 42 male Wistar rats were used. Recombinant adeno-associated viruses were used to label and regulate the activity of dopamine (DA) neurons. The rat model was established by complete Freund's adjuvant (CFA). Lower limb electroacupuncture (LEA) was applied to the ST36 and BL60 acupoints. In addition, LEA + scalp EA (SEA) was given using the GV20 and GV24+ acupoints besides ST36 and BL60. To evaluate the pain threshold, we measured 50% paw withdrawal thresholds and thermal paw withdrawal latencies. Negative emotions were evaluated through the open field test, marble-burying test, sucrose preference test, and forced swimming test. Moreover, the conditional place preference test was conducted to measure the reward behavior in response to pain relief. Immunofluorescence staining, Western blotting, and qPCR were used to detect the activity of the VTADA-NAc reward circuit. Result The injection of CFA significantly lowered the pain threshold. As the pain persisted, the anxiety and depression-like behaviors escalated while the response to reward reduced. Meanwhile, the VTADA-NAc pathway was suppressed with pain chronification. However, activating DA neurons in VTA attenuated the effects induced by CFA. LEA could relieve chronic pain, negative emotions, and reward disorders, while also activating the VTADA-NAc pathway. In addition, LEA + SEA exhibited a more pronounced effect compared with LEA alone. Nevertheless, chemogenetic inhibition of DA neurons decreased the efficacy of LEA + SEA in the treatment of chronic pain and associated comorbidities. Conclusion Adding SEA to conventional LEA effectively alleviates negative emotions and chronic pain, potentially due to the activation of the VTADA-NAc reward neural circuit. Thus, LEA + SEA is a more effective treatment for hyperalgesia and associated negative emotions compared with LEA alone.
Collapse
Affiliation(s)
- Yanan Yang
- Department of Traditional Chinese Medicine, China Resources & Wugang General Hospital, Wuhan, China
| | - Xiali Wei
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ye Zhu
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shaohui Jia
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
Wayne CR, Karam AM, McInnis AL, Arms CM, Kaller MD, Maruska KP. Impacts of repeated social defeat on behavior and the brain in a cichlid fish. J Exp Biol 2023; 226:jeb246322. [PMID: 37909345 DOI: 10.1242/jeb.246322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
Social defeat is a powerful experience leading to drastic changes in physiology and behavior, many of which are negative. For example, repeated social defeat in vertebrates results in reduced reproductive success, sickness and behavioral abnormalities that threaten individual survival and species persistence. However, little is known about what neural mechanisms are involved in determining whether an individual is resilient or susceptible to repeated social defeat stress. It also remains unknown whether exclusive use of reactive behaviors after repeated social defeat is maintained over time and impacts future behaviors during subsequent contests. We used a resident-intruder experiment in the African cichlid fish Astatotilapia burtoni to investigate the behavior and neural correlates of these two opposing groups. Behavior was quantified by watching fish during defeat trials and used to distinguish resilient and susceptible individuals. Both resilient and susceptible fish started with searching and freezing behaviors, with searching decreasing and freezing increasing after repeated social defeat. After a 4 day break period, resilient fish used both searching and freezing behaviors during a social defeat encounter with a new resident, while susceptible fish almost exclusively used freezing behaviors. By quantifying neural activation using pS6 in socially relevant brain regions, we identified differential neural activation patterns associated with resilient and susceptible fish and found nuclei that co-varied and may represent functional networks. These data provide the first evidence of specific conserved brain networks underlying social stress resilience and susceptibility in fishes.
Collapse
Affiliation(s)
- C Rose Wayne
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Bldg, Baton Rouge, LA 70803, USA
| | - Ava M Karam
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Bldg, Baton Rouge, LA 70803, USA
| | - Alora L McInnis
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Bldg, Baton Rouge, LA 70803, USA
| | - Catherine M Arms
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Bldg, Baton Rouge, LA 70803, USA
| | - Michael D Kaller
- School of Renewable Natural Resources, Louisiana State University Agricultural Center, Baton Rouge, LA 70803, USA
| | - Karen P Maruska
- Department of Biological Sciences, Louisiana State University, 202 Life Sciences Bldg, Baton Rouge, LA 70803, USA
| |
Collapse
|
41
|
El Mansari M, Hamoudeh R, Daniels S, Blier P. Wistar Kyoto rats exhibit decreased serotonin neuronal firing and increased norepinephrine burst activity but dampened hippocampal α 2-adrenoceptor sensitivity. J Psychopharmacol 2023; 37:1105-1115. [PMID: 37942525 DOI: 10.1177/02698811231209235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
BACKGROUND Wistar Kyoto (WKY) rats manifest abnormalities in the function of monoamine receptors and transporters, as well as levels of these neurotransmitters in the brain. The present study assessed alterations in the firing activity of serotonin (5-hydroxytryptamine [5-HT]), norepinephrine (NE), and dopamine (DA) neurons, as well as the activity of 5-HT and NE receptors and transporters in the hippocampus. METHODS In vivo electrophysiological recordings were conducted in male WKY and Wistar rats. Extracellular single-unit recordings of 5-HT, NE, and DA neurons were performed. Recordings of pyramidal neurons were conducted in the medial prefrontal cortex (mPFC) and the hippocampus, where direct application of 5-HT and NE by iontophoresis was also carried out. RESULTS The mean firing rate of 5-HT neurons was significantly decreased in WKY compared to Wistar rats. The burst activity of NE neurons was significantly increased in WKY, while their mean firing activity was not changed. There was no alteration in the firing, burst, and population activity of DA neurons in WKY animals. In the hippocampus, a decrease in sensitivity of α2-adrenoceptors, but not 5-HT receptors, was observed. There was, however, no change in the activity of 5-HT and NE transporters. The firing activity of mPFC pyramidal neurons was similar in WKY versus Wistar rats. CONCLUSION In WKY rats, there was a decrease in the firing activity of 5-HT neurons. There was also an enhanced burst activity of NE neurons, accompanied by a reduction in sensitivity of the α2-adrenoceptor in the hippocampus, inferring a decrease in NE transmission.
Collapse
Affiliation(s)
- Mostafa El Mansari
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Rami Hamoudeh
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Stephen Daniels
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Pierre Blier
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| |
Collapse
|
42
|
Heberden C, Maximin E, Rabot S, Naudon L. Male mice engaging differently in emotional eating present distinct plasmatic and neurological profiles. Nutr Neurosci 2023; 26:1034-1044. [PMID: 36154930 DOI: 10.1080/1028415x.2022.2122137] [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: 10/14/2022]
Abstract
Objective: Stressed individuals tend to turn to calorie-rich food, also known as 'comfort food' for the temporary relief it provides. The emotional eating drive is highly variable among subjects. Using a rodent model, we explored the plasmatic and neurobiological differences between 'high and low emotional eaters' (HEE and LEE).Methods: 40 male mice were exposed for 5 weeks to a protocol of unpredictable chronic mild stress. Every 3 or 4 days, they were submitted to a 1-h restraint stress, immediately followed by a 3-h period during which a choice between chow and chocolate sweet cereals was proposed. The dietary intake was measured by weighing. Plasmatic and neurobiological characteristics were compared in mice displaying high vs low intakes.Results: Out of 40 mice, 8 were considered as HEE because of their high post-stress eating score, and 8 as LEE because of their consistent low intake. LEE displayed higher plasma corticosterone and lower levels of NPY than HEE, but acylated and total ghrelin were similar in both groups. In the brain, the abundance of NPY neurons in the arcuate nucleus of the hypothalamus was similar in both groups, but was higher in the ventral hippocampus and the basal lateral amygdala of LEE. The lateral hypothalamus LEE had also more orexin (OX) positive neurons. Both NPY and OX are orexigenic peptides and mood regulators.Discussion: Emotional eating difference was reflected in plasma and brain structures implicated in emotion and eating regulation. These results concur with the psychological side of food consumption.
Collapse
Affiliation(s)
- Christine Heberden
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Elise Maximin
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Sylvie Rabot
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| | - Laurent Naudon
- INRAE, AgroParisTech, CNRS, Micalis Institute, Université Paris-Saclay Jouy-en-Josas, France
| |
Collapse
|
43
|
Cai M, Zhu Y, Shanley MR, Morel C, Ku SM, Zhang H, Shen Y, Friedman AK, Han MH. HCN channel inhibitor induces ketamine-like rapid and sustained antidepressant effects in chronic social defeat stress model. Neurobiol Stress 2023; 26:100565. [PMID: 37664876 PMCID: PMC10468802 DOI: 10.1016/j.ynstr.2023.100565] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
Repeated, long-term (weeks to months) exposure to standard antidepressant medications is required to achieve treatment efficacy. In contrast, acute ketamine quickly improves mood for an extended time. Recent work implicates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are involved in mediating ketamine's antidepressant effects. In this study, we directly targeted HCN channels and achieved ketamine-like rapid and sustained antidepressant efficacy. Our in vitro electrophysiological recordings first showed that HCN inhibitor DK-AH 269 (also called cilobradine) decreased the pathological HCN-mediated current (Ih) and abnormal hyperactivity of ventral tegmental area (VTA) dopamine (DA) neurons in a depressive-like model produced by chronic social defeat stress (CSDS). Our in vivo studies further showed that acute intra-VTA or acute systemic administration of DK-AH 269 normalized social behavior and rescued sucrose preference in CSDS-susceptible mice. The single-dose of DK-AH 269, both by intra-VTA microinfusion and intraperitoneal (ip) approaches, could produce an extended 13-day duration of antidepressant-like efficacy. Animals treated with acute DK-AH 269 spent less time immobile than vehicle-treated mice during forced swim test. A social behavioral reversal lasted up to 13 days following the acute DK-AH 269 ip injection, and this rapid and sustained antidepressant-like response is paralleled with a single-dose treatment of ketamine. This study provides a novel ion channel target for acutely acting, long-lasting antidepressant-like effects.
Collapse
Affiliation(s)
- Min Cai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yingbo Zhu
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- China Shenzhen Naowunao Network Technology Co.,Ltd., Shenzhen, Guangdong, China
| | - Mary Regis Shanley
- Department of Biological Sciences, Hunter College, Biology and Biochemistry PhD Program, Graduate Center, The City University of New York, New York, NY, USA
| | - Carole Morel
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stacy M. Ku
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hongxing Zhang
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuan Shen
- Anesthesia and Brain Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Allyson K. Friedman
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, China
| |
Collapse
|
44
|
Kokorikou DS, Sarigiannidis I, Fiore VG, Parkin B, Hopkins A, El-Deredy W, Dilley L, Moutoussis M. Testing hypotheses about the harm that capitalism causes to the mind and brain: a theoretical framework for neuroscience research. FRONTIERS IN SOCIOLOGY 2023; 8:1030115. [PMID: 37404338 PMCID: PMC10315660 DOI: 10.3389/fsoc.2023.1030115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023]
Abstract
In this paper, we will attempt to outline the key ideas of a theoretical framework for neuroscience research that reflects critically on the neoliberal capitalist context. We argue that neuroscience can and should illuminate the effects of neoliberal capitalism on the brains and minds of the population living under such socioeconomic systems. Firstly, we review the available empirical research indicating that the socio-economic environment is harmful to minds and brains. We, then, describe the effects of the capitalist context on neuroscience itself by presenting how it has been influenced historically. In order to set out a theoretical framework that can generate neuroscientific hypotheses with regards to the effects of the capitalist context on brains and minds, we suggest a categorization of the effects, namely deprivation, isolation and intersectional effects. We also argue in favor of a neurodiversity perspective [as opposed to the dominant model of conceptualizing neural (mal-)functioning] and for a perspective that takes into account brain plasticity and potential for change and adaptation. Lastly, we discuss the specific needs for future research as well as a frame for post-capitalist research.
Collapse
Affiliation(s)
- Danae S. Kokorikou
- Psychoanalysis Unit, Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom
| | - Ioannis Sarigiannidis
- Institute of Cognitive Neuroscience, University College London, London, United Kingdom
| | - Vincenzo G. Fiore
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Beth Parkin
- Department of Psychology, School of Social Sciences, University of Westminster, London, United Kingdom
| | - Alexandra Hopkins
- Department of Psychology, Royal Holloway, University of London, London, United Kingdom
| | - Wael El-Deredy
- Centro de Investigación y Desarrollo en Ingeniería en Salud, Universidad de Valparaíso, Valparaíso, Chile
| | - Laura Dilley
- Department of Communicative Sciences and Disorders, Michigan State University, East Lansing, MI, United States
| | - Michael Moutoussis
- Wellcome Centre for Human Neuroimaging, University College London, London, United Kingdom
- Max Planck University College London Centre for Computational Psychiatry and Ageing Research, London, United Kingdom
| |
Collapse
|
45
|
Khairuddin S, Lim WL, Aquili L, Tsui KC, Tse ACK, Jayalath S, Varma R, Sharp T, Benazzouz A, Steinbusch H, Blokland A, Temel Y, Lim LW. Prelimbic Cortical Stimulation Induces Antidepressant-like Responses through Dopaminergic-Dependent and -Independent Mechanisms. Cells 2023; 12:1449. [PMID: 37296570 PMCID: PMC10253143 DOI: 10.3390/cells12111449] [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: 04/05/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
High-frequency stimulation (HFS) is a promising therapy for patients with depression. However, the mechanisms underlying the HFS-induced antidepressant-like effects on susceptibility and resilience to depressive-like behaviors remain obscure. Given that dopaminergic neurotransmission has been found to be disrupted in depression, we investigated the dopamine(DA)-dependent mechanism of the antidepressant-like effects of HFS of the prelimbic cortex (HFS PrL). We performed HFS PrL in a rat model of mild chronic unpredictable stress (CUS) together with 6-hydroxydopamine lesioning in the dorsal raphe nucleus (DRN) and ventral tegmental area (VTA). Animals were assessed for anxiety, anhedonia, and behavioral despair. We also examined levels of corticosterone, hippocampal neurotransmitters, neuroplasticity-related proteins, and morphological changes in dopaminergic neurons. We found 54.3% of CUS animals exhibited decreased sucrose consumption and were designated as CUS-susceptible, while the others were designated CUS-resilient. HFS PrL in both the CUS-susceptible and CUS-resilient animals significantly increased hedonia, reduced anxiety, decreased forced swim immobility, enhanced hippocampal DA and serotonin levels, and reduced corticosterone levels when compared with the respective sham groups. The hedonic-like effects were abolished in both DRN- and VTA-lesioned groups, suggesting the effects of HFS PrL are DA-dependent. Interestingly, VTA-lesioned sham animals had increased anxiety and forced swim immobility, which was reversed by HFS PrL. The VTA-lesioned HFS PrL animals also had elevated DA levels, and reduced p-p38 MAPK and NF-κB levels when compared to VTA-lesioned sham animals. These findings suggest that HFS PrL in stressed animals leads to profound antidepressant-like responses possibly through both DA-dependent and -independent mechanisms.
Collapse
Affiliation(s)
- Sharafuddin Khairuddin
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wei Ling Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Biological Sciences, Sunway University, Bandar Sunway, Petaling Jaya 47500, Malaysia
| | - Luca Aquili
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Discipline of Psychology, College of Health and Education, Murdoch University, Perth 6150, Australia
| | - Ka Chun Tsui
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anna Chung-Kwan Tse
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shehani Jayalath
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ruhani Varma
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Abdelhamid Benazzouz
- CNRS UMR5293, Institute of Neurodegenerative Diseases, University de Bordeaux, 33000 Bordeaux, France
| | - Harry Steinbusch
- Department of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Arjan Blokland
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neuroscience, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Neurosurgery, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Department of Biological Sciences, Sunway University, Bandar Sunway, Petaling Jaya 47500, Malaysia
| |
Collapse
|
46
|
Pinares-Garcia P, Spyrou J, McKenzie CE, Forster IC, Soh MS, Mohamed Syazwan E, Atif M, Reid CA. Antidepressant-like activity of a brain penetrant HCN channel inhibitor in mice. Front Pharmacol 2023; 14:1159527. [PMID: 37234718 PMCID: PMC10206048 DOI: 10.3389/fphar.2023.1159527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Changes in Hyperpolarization-Activated Cyclic Nucleotide-Gated (HCN) channel function have been linked to depressive-like traits, making them potential drug targets. However, there is currently no peer-reviewed data supporting the use of a small molecule modulator of HCN channels in depression treatment. Org 34167, a benzisoxazole derivative, has been patented for the treatment of depression and progressed to Phase I trials. In the current study, we analysed the biophysical effects of Org 34167 on HCN channels in stably transfected human embryonic kidney 293 (HEK293) cells and mouse layer V neurons using patch-clamp electrophysiology, and we utilised three high-throughput screens for depressive-like behaviour to assess the activity of Org 34167 in mice. The impact of Org 34167 on locomotion and coordination were measured by performing rotarod and ledged beam tests. Org 34167 is a broad-spectrum inhibitor of HCN channels, slowing activation and causing a hyperpolarising shift in voltage-dependence of activation. It also reduced I h-mediated sag in mouse neurons. Org 34167 (0.5 mg/kg) reduced marble burying and increased the time spent mobile in the Porsolt swim and tail suspension tests in both male and female BALB/c mice, suggesting reduced depressive-like behaviour. Although no adverse effects were seen at 0.5 mg/kg, an increase in dose to 1 mg/kg resulted in visible tremors and impaired locomotion and coordination. These data support the premise that HCN channels are valid targets for anti-depressive drugs albeit with a narrow therapeutic index. Drugs with higher HCN subtype selectivity are needed to establish if a wider therapeutic window can be obtained.
Collapse
Affiliation(s)
- Paulo Pinares-Garcia
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - James Spyrou
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Chaseley E. McKenzie
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ian C. Forster
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Ming S. Soh
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Erlina Mohamed Syazwan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Mohammed Atif
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Christopher A. Reid
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Heidelberg, VIC, Australia
| |
Collapse
|
47
|
Papp M, Gruca P, Litwa E, Lason M, Willner P. Optogenetic stimulation of transmission from prelimbic cortex to nucleus accumbens core overcomes resistance to venlafaxine in an animal model of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110715. [PMID: 36610613 DOI: 10.1016/j.pnpbp.2023.110715] [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: 07/17/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
BACKGROUND Our earlier study demonstrated that repeated optogenetic stimulation of afferents from ventral hippocampus (vHIP) to the prelimbic region of medial prefrontal cortex (mPFC) overcame resistance to antidepressant treatment in Wistar-Kyoto (WKY) rats. These results suggested that antidepressant resistance may result from an insufficiency of transmission from vHIP to mPFC. Here we examined whether similar effects can be elicited from major output of mPFC; the pathway from to nucleus accumbens core (NAc). METHOD WKY rats were subjected to Chronic Mild Stress and were used in two sets of experiments: 1) they were treated acutely with optogenetic stimulation of afferents to NAc core originating from the mPFC, and 2) they were treated with chronic (5 weeks) venlafaxine (10 mg/kg) and/or repeated (once weekly) optogenetic stimulation of afferents to NAc originating from either mPFC or vHIP. RESULTS Chronic mild stress procedure decreased sucrose intake, open arm entries on elevated plus maze, and novel object recognition test. Acute optogenetic stimulation of the mPFC-NAc and vHIP-NAc pathways had no effect in sucrose or plus maze tests, but increased object recognition. Neither venlafaxine nor mPFC-NAc optogenetic stimulation alone was effective in reversing the effects of CMS, but the combination of chronic antidepressant and repeated optogenetic stimulation improved behaviour on all three measures. CONCLUSIONS The synergism between venlafaxine and mPFC-NAc optogenetic stimulation supports the hypothesis that the mechanisms of non-responsiveness of WKY rats involves a failure of antidepressant treatment to restore transmission in the mPFC-NAc pathway. Together with earlier results, this implicates insufficiency in a vHIP-mPFC-NAc circuit in non-responsiveness to antidepressant drugs.
Collapse
Affiliation(s)
- Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Paul Willner
- Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
48
|
Cardona-Acosta AM, Bolaños-Guzmán CA. Role of the mesolimbic dopamine pathway in the antidepressant effects of ketamine. Neuropharmacology 2023; 225:109374. [PMID: 36516891 PMCID: PMC9839658 DOI: 10.1016/j.neuropharm.2022.109374] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/27/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
Depression is a complex and highly heterogeneous disorder which diagnosis is based on an exceedingly variable set of clinical symptoms. Current treatments focus almost exclusively on the manipulation of monoamine neurotransmitter systems, but despite considerable efforts, these remain inadequate for a significant proportion of those afflicted by the disorder. The emergence of racemic (R, S)-ketamine as a fast-acting antidepressant has provided an exciting new path for the study of major depressive disorder (MDD) and the search for better therapeutics for its treatment. Previous work suggested that ketamine's mechanism of action is primarily mediated via blockaded of N-methyl-d-aspartate (NMDA) receptors, however, this is an area of active research and clinical and preclinical evidence now indicate that ketamine acts on multiple systems. The last couple of decades have cemented the mesolimbic dopamine reward pathway's involvement in the pathogenesis of MDD and related mood disorders. Exposure to negative stress dysregulates dopamine neuronal activity disrupting reward and motivational processes resulting in anhedonia (lack of pleasure), a hallmark symptom of depression. Although the mechanism(s) underlying ketamine's antidepressant activity continue to be elucidated, current evidence indicate that its therapeutic effects are mediated, at least in part, via long-lasting synaptic changes and subsequent molecular adaptations in brain regions within the mesolimbic dopamine system. Notwithstanding, ketamine is a drug of abuse, and this liability may pose limitations for long term use as an antidepressant. This review outlines the current knowledge of ketamine's actions within the mesolimbic dopamine system and its abuse potential. This article is part of the Special Issue on 'Ketamine and its Metabolites'.
Collapse
Affiliation(s)
- Astrid M Cardona-Acosta
- Department of Psychological and Brain Sciences and Program in Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Carlos A Bolaños-Guzmán
- Department of Psychological and Brain Sciences and Program in Neuroscience, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
49
|
The laterodorsal tegmentum-ventral tegmental area circuit controls depression-like behaviors by activating ErbB4 in DA neurons. Mol Psychiatry 2023; 28:1027-1045. [PMID: 33990773 PMCID: PMC8590712 DOI: 10.1038/s41380-021-01137-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 04/08/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023]
Abstract
Dopamine (DA) neurons in the ventral tegmental area (VTA) are critical to coping with stress. However, molecular mechanisms regulating their activity and stress-induced depression were not well understood. We found that the receptor tyrosine kinase ErbB4 in VTA was activated in stress-susceptible mice. Deleting ErbB4 in VTA or in DA neurons, or chemical genetic inhibition of ErbB4 kinase activity in VTA suppressed the development of chronic social defeat stress (CSDS)-induced depression-like behaviors. ErbB4 activation required the expression of NRG1 in the laterodorsal tegmentum (LDTg); LDTg-specific deletion of NRG1 inhibited depression-like behaviors. NRG1 and ErbB4 suppressed potassium currents of VTA DA neurons and increased their firing activity. Finally, we showed that acute inhibition of ErbB4 after stress attenuated DA neuron hyperactivity and expression of depression-like behaviors. Together, these observations demonstrate a critical role of NRG1-ErbB4 signaling in regulating depression-like behaviors and identify an unexpected mechanism by which the LDTg-VTA circuit regulates the activity of DA neurons.
Collapse
|
50
|
Mood and behavior regulation: interaction of lithium and dopaminergic system. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023:10.1007/s00210-023-02437-1. [PMID: 36843130 DOI: 10.1007/s00210-023-02437-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/20/2023] [Indexed: 02/28/2023]
Abstract
Lithium is one of the most effect mood-stabilizing drugs prescribed especially for bipolar disorder. Lithium has wide range effects on different molecular factors and neural transmission including dopaminergic signaling. On the other hand, mesolimbic and mesocortical dopaminergic signaling is significantly involved in the pathophysiology of neuropsychiatric disorders. This review article aims to study lithium therapeutic mechanisms, dopaminergic signaling, and the interaction of lithium and dopamine. We concluded that acute and chronic lithium treatments often reduce dopamine synthesis and level in the brain. However, some studies have reported conflicting results following lithium treatment, especially chronic treatment. The dosage, duration, and type of lithium administration, and the brain region selected for measuring dopamine level were not significant differences in different chronic treatments used in previous studies. It was suggested that lithium has various mechanisms affecting dopaminergic signaling and mood, and that many molecular factors can be involved, including brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), β-catenin, protein kinase B (Akt), and glycogen synthase kinase-3 beta (GSK-3β). Thus, molecular effects of lithium can be the most important mechanisms of lithium that also alter neural transmissions including dopaminergic signaling in mesolimbic and mesocortical pathways.
Collapse
|