1
|
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
|
2
|
Maran R, Müller EJ, Fulcher BD. Analyzing the brain's dynamic response to targeted stimulation using generative modeling. Netw Neurosci 2025; 9:237-258. [PMID: 40161996 PMCID: PMC11949581 DOI: 10.1162/netn_a_00433] [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: 08/05/2024] [Accepted: 11/19/2024] [Indexed: 04/02/2025] Open
Abstract
Generative models of brain activity have been instrumental in testing hypothesized mechanisms underlying brain dynamics against experimental datasets. Beyond capturing the key mechanisms underlying spontaneous brain dynamics, these models hold an exciting potential for understanding the mechanisms underlying the dynamics evoked by targeted brain stimulation techniques. This paper delves into this emerging application, using concepts from dynamical systems theory to argue that the stimulus-evoked dynamics in such experiments may be shaped by new types of mechanisms distinct from those that dominate spontaneous dynamics. We review and discuss (a) the targeted experimental techniques across spatial scales that can both perturb the brain to novel states and resolve its relaxation trajectory back to spontaneous dynamics and (b) how we can understand these dynamics in terms of mechanisms using physiological, phenomenological, and data-driven models. A tight integration of targeted stimulation experiments with generative quantitative modeling provides an important opportunity to uncover novel mechanisms of brain dynamics that are difficult to detect in spontaneous settings.
Collapse
Affiliation(s)
- Rishikesan Maran
- School of Physics, University of Sydney, Camperdown Campus, Sydney, NSW, Australia
| | - Eli J. Müller
- School of Physics, University of Sydney, Camperdown Campus, Sydney, NSW, Australia
| | - Ben D. Fulcher
- School of Physics, University of Sydney, Camperdown Campus, Sydney, NSW, Australia
| |
Collapse
|
3
|
Zhu L, Wang Y, Wu X, Wu G, Zhang G, Liu C, Zhang S. Protein design accelerates the development and application of optogenetic tools. Comput Struct Biotechnol J 2025; 27:717-732. [PMID: 40092664 PMCID: PMC11908464 DOI: 10.1016/j.csbj.2025.02.014] [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: 12/04/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Optogenetics has substantially enhanced our understanding of biological processes by enabling high-precision tracking and manipulation of individual cells. It relies on photosensitive proteins to monitor and control cellular activities, thereby paving the way for significant advancements in complex system research. Photosensitive proteins play a vital role in the development of optogenetics, facilitating the establishment of cutting-edge methods. Recent breakthroughs in protein design have opened up opportunities to develop protein-based tools that can precisely manipulate and monitor cellular activities. These advancements will significantly accelerate the development and application of optogenetic tools. This article emphasizes the pivotal role of protein design in the development of optogenetic tools, offering insights into potential future directions. We begin by providing an introduction to the historical development and fundamental principles of optogenetics, followed by an exploration of the operational mechanisms of key photosensitive domains, which includes clarifying the conformational changes they undergo in response to light, such as allosteric modulation and dimerization processes. Building on this foundation, we reveal the development of protein design tools that will enable the creation of even more sophisticated optogenetic techniques.
Collapse
Affiliation(s)
| | | | - Xiaomin Wu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Guohua Wu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Guohao Zhang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Chuanyang Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Shaowei Zhang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan 410073, China
| |
Collapse
|
4
|
Kuga N, Sasaki T. Memory-related neurophysiological mechanisms in the hippocampus underlying stress susceptibility. Neurosci Res 2025; 211:3-9. [PMID: 35931215 DOI: 10.1016/j.neures.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 07/20/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022]
Abstract
Stress-induced psychiatric symptoms, such as increased anxiety, decreased sociality, and depression, differ considerably across individuals. The cognitive model of depression proposes that biased negative memory is a crucial determinant in the development of mental stress-induced disorders. Accumulating evidence from both clinical and animal studies has demonstrated that such biased memory processing could be triggered by the hippocampus, a region well known to be involved in declarative memories. This review mainly describes how memory-related neurophysiological mechanisms in the hippocampus and their interactions with other related brain regions are involved in the regulation of stress susceptibility and discusses potential interventions to prevent and treat stress-related psychiatric symptoms. Further neurophysiological insights based on memory mechanisms are expected to devise personalized prevention and therapy to confer stress resilience.
Collapse
Affiliation(s)
- Nahoko Kuga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan
| | - Takuya Sasaki
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-Ku, Sendai 980-8578, Japan.
| |
Collapse
|
5
|
Reimer AE, Dastin-van Rijn EM, Kim J, Mensinger ME, Sachse EM, Wald A, Hoskins E, Singh K, Alpers A, Cooper D, Lo MC, de Oliveira AR, Simandl G, Stephenson N, Widge AS. Striatal stimulation enhances cognitive control and evidence processing in rodents and humans. Sci Transl Med 2024; 16:eadp1723. [PMID: 39693410 DOI: 10.1126/scitranslmed.adp1723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/05/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
Brain disorders, in particular mental disorders, might be effectively treated by direct electrical brain stimulation, but clinical progress requires understanding of therapeutic mechanisms. Animal models have not helped, because there are no direct animal models of mental illness. Here, we propose a potential path past this roadblock, by leveraging a common ingredient of most mental disorders: impaired cognitive control. We previously showed that deep brain stimulation (DBS) improves cognitive control in humans. We now reverse translate that result using a set-shifting task in rats. DBS-like stimulation of the midstriatum improved reaction times without affecting accuracy, mirroring our human findings. Impulsivity, motivation, locomotor, and learning effects were ruled out through companion tasks and model-based analyses. To identify the specific cognitive processes affected, we applied reinforcement learning drift-diffusion modeling. This approach revealed that DBS-like stimulation enhanced evidence accumulation rates and lowered decision thresholds, improving domain-general cognitive control. Reanalysis of prior human data showed that the same mechanism applies in humans. This reverse/forward translational model could have near-term implications for clinical DBS practice and future trial design.
Collapse
Affiliation(s)
- Adriano E Reimer
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Evan M Dastin-van Rijn
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Jaejoong Kim
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Megan E Mensinger
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Elizabeth M Sachse
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Aaron Wald
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Eric Hoskins
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Kartikeya Singh
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Abigail Alpers
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Dawson Cooper
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Meng-Chen Lo
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | | | - Gregory Simandl
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Nathaniel Stephenson
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| | - Alik S Widge
- Department of Psychiatry & Behavioral Sciences, University of Minnesota, Minnesota, MN 55454, USA
| |
Collapse
|
6
|
Paoletti I, Coccurello R. Irisin: A Multifaceted Hormone Bridging Exercise and Disease Pathophysiology. Int J Mol Sci 2024; 25:13480. [PMID: 39769243 PMCID: PMC11676223 DOI: 10.3390/ijms252413480] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
The fibronectin domain-containing protein 5 (FNDC5), or irisin, is an adipo-myokine hormone produced during exercise, which shows therapeutic potential for conditions like metabolic disorders, osteoporosis, sarcopenia, obesity, type 2 diabetes, and neurodegenerative diseases, including Alzheimer's disease (AD). This review explores its potential across various pathophysiological processes that are often considered independent. Elevated in healthy states but reduced in diseases, irisin improves muscle-adipose communication, insulin sensitivity, and metabolic balance by enhancing mitochondrial function and reducing oxidative stress. It promotes osteogenesis and mitigates bone loss in osteoporosis and sarcopenia. Irisin exhibits anti-inflammatory effects by inhibiting NF-κB signaling and countering insulin resistance. In the brain, it reduces amyloid-β toxicity, inflammation, and oxidative stress, enhancing brain-derived neurotrophic factor (BDNF) signaling, which improves cognition and synaptic health in AD models. It also regulates dopamine pathways, potentially alleviating neuropsychiatric symptoms like depression and apathy. By linking physical activity to systemic health, irisin emphasizes its role in the muscle-bone-brain axis. Its multifaceted benefits highlight its potential as a therapeutic target for AD and related disorders, with applications in prevention, in treatment, and as a complement to exercise strategies.
Collapse
Affiliation(s)
- Ilaria Paoletti
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Research Council (C.N.R.), 00185 Rome, Italy
| |
Collapse
|
7
|
Hing B, Mitchell SB, Filali Y, Eberle M, Hultman I, Matkovich M, Kasturirangan M, Johnson M, Wyche W, Jimenez A, Velamuri R, Ghumman M, Wickramasinghe H, Christian O, Srivastava S, Hultman R. Transcriptomic Evaluation of a Stress Vulnerability Network Using Single-Cell RNA Sequencing in Mouse Prefrontal Cortex. Biol Psychiatry 2024; 96:886-899. [PMID: 38866174 PMCID: PMC11524784 DOI: 10.1016/j.biopsych.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 04/24/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Increased vulnerability to stress is a major risk factor for several mood disorders, including major depressive disorder. Although cellular and molecular mechanisms associated with depressive behaviors following stress have been identified, little is known about the mechanisms that confer the vulnerability that predisposes individuals to future damage from chronic stress. METHODS We used multisite in vivo neurophysiology in freely behaving male and female C57BL/6 mice (n = 12) to measure electrical brain network activity previously identified as indicating a latent stress vulnerability brain state. We combined this neurophysiological approach with single-cell RNA sequencing of the prefrontal cortex to identify distinct transcriptomic differences between groups of mice with inherent high and low stress vulnerability. RESULTS We identified hundreds of differentially expressed genes (padjusted < .05) across 5 major cell types in animals with high and low stress vulnerability brain network activity. This unique analysis revealed that GABAergic (gamma-aminobutyric acidergic) neuron gene expression contributed most to the network activity of the stress vulnerability brain state. Upregulation of mitochondrial and metabolic pathways also distinguished high and low vulnerability brain states, especially in inhibitory neurons. Importantly, genes that were differentially regulated with vulnerability network activity significantly overlapped (above chance) with those identified by genome-wide association studies as having single nucleotide polymorphisms significantly associated with depression as well as genes more highly expressed in postmortem prefrontal cortex of patients with major depressive disorder. CONCLUSIONS This is the first study to identify cell types and genes involved in a latent stress vulnerability state in the brain.
Collapse
Affiliation(s)
- Benjamin Hing
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Sara B Mitchell
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Yassine Filali
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Maureen Eberle
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Ian Hultman
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, Iowa
| | - Molly Matkovich
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | | | - Micah Johnson
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa
| | - Whitney Wyche
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Alli Jimenez
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Radha Velamuri
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Mahnoor Ghumman
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Himali Wickramasinghe
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Olivia Christian
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
| | - Sanvesh Srivastava
- Department of Statistics and Actuarial Science, University of Iowa, Iowa City, Iowa
| | - Rainbo Hultman
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; Department of Psychiatry, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
8
|
Deli A, Green AL. Deep Brain Stimulation for Consciousness Disorders; Technical and Ethical Considerations. NEUROETHICS-NETH 2024; 17:35. [PMID: 39091894 PMCID: PMC11289033 DOI: 10.1007/s12152-024-09570-5] [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: 12/31/2023] [Accepted: 07/10/2024] [Indexed: 08/04/2024]
Abstract
Disorders of Consciousness (DoC) result in profound functional impairment, adversely affecting the lives of a predominantly younger patient population. Currently, effective treatment options for those who have reached chronicity (prolonged symptom duration over 4 weeks) are extremely limited, with the majority of such cases facing life-long dependence on carers and a poor quality of life. Here we briefly review the current evidence on caseload, diagnostic and management options in the United Kingdom (UK), United States of America (USA) and the European Union (EU). We identify key differences as well as similarities in these approaches across respective healthcare systems, highlighting unmet needs in this population. We subsequently present past efforts and the most recent advances in the field of surgical modulation of consciousness through implantable neurostimulation systems. We examine the ethical dilemmas that such a treatment approach may pose, proposing mediating solutions and methodological adjustments to address these concerns. Overall, we argue that there is a strong case for the utilisation of deep brain stimulation (DBS) in the DoC patient cohort. This is based on both promising results of recent clinical trials as well as technological developments. We propose a revitalization of surgical neuromodulation for DoC with a multicenter, multidisciplinary approach and strict monitoring guidelines, in order to not only advance treatment options but also ensure the safeguarding of patients' welfare and dignity.
Collapse
Affiliation(s)
- Alceste Deli
- Nuffield Department of Surgical Sciences and Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alexander L. Green
- Nuffield Department of Surgical Sciences and Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Zhang Y, Jiang Y, Yu Z, Li Y, Lin X, Weng Y, Guo Z, Hu H, Shao W, Yu G, Zheng F, Cai P, Li H, Wu S. VGluT2 neuron subtypes in the paraventricular thalamic nucleus regulate depression in paraquat-induced Parkinson's disease. JOURNAL OF HAZARDOUS MATERIALS 2024; 472:134559. [PMID: 38735189 DOI: 10.1016/j.jhazmat.2024.134559] [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: 01/22/2024] [Revised: 04/14/2024] [Accepted: 05/05/2024] [Indexed: 05/14/2024]
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease and approximately one third of patients with PD are estimated to experience depression. Paraquat (PQ) is the most widely used herbicide worldwide and PQ exposure is reported to induce PD with depression. However, the specific brain region and neural networks underlying the etiology of depression in PD, especially in the PQ-induced model, have not yet been elucidated. Here, we report that the VGluT2-positive glutamatergic neurons in the paraventricular thalamic nucleus (PVT) promote depression in the PQ-induced PD mouse model. Our results show that PVTVGluT2 neurons are activated by PQ and their activation increases the susceptibility to depression in PD mice. Conversely, inhibition of PVTVGluT2 neurons reversed the depressive-behavioral changes induced by PQ. Similar to the effects of intervention the soma of PVTVGluT2 neurons, stimulation of their projections into the central amygdaloid nucleus (CeA) also strongly influenced depression in PD mice. PQ induced malfunctioning of the glutamate system and changes in the dendritic and synaptic morphology in the CeA through its role on PVTVGluT2 neuronal activation. In summary, our results demonstrate that PVTVGluT2 neurons are key neuronal subtypes for depression in PQ-induced PD and promote depression processes through the PVTVGluT2-CeA pathway.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yihua Jiang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Zhen Yu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yinhan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Xinpei Lin
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Yali Weng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Zhenkun Guo
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Hong Hu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Wenya Shao
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Guangxia Yu
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Fuli Zheng
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Ping Cai
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China
| | - Huangyuan Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China; The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| | - Siying Wu
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Fujian Provincial Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350122, China; Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
10
|
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
|
11
|
Fermin ASR, Sasaoka T, Maekawa T, Ono K, Chan HL, Yamawaki S. Insula-cortico-subcortical networks predict interoceptive awareness and stress resilience. Asian J Psychiatr 2024; 95:103991. [PMID: 38484483 DOI: 10.1016/j.ajp.2024.103991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/25/2024] [Accepted: 02/28/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Interoception, the neural sensing of visceral signals, and interoceptive awareness (IA), the conscious perception of interoception, are crucial for life survival functions and mental health. Resilience, the capacity to overcome adversity, has been associated with reduced interoceptive disturbances. Here, we sought evidence for our Insula Modular Active Control (IMAC) model that suggest that the insula, a brain region specialized in the processing of interoceptive information, realizes IA and contributes to resilience and mental health via cortico-subcortical connections. METHODS 64 healthy participants (32 females; ages 18-34 years) answered questionnaires that assess IA and resilience. Mental health was evaluated with the Beck Depression Inventory II that assesses depressive mood. Participants also underwent a 15 minute resting-state functional resonance imaging session. Pearson correlations and mediation analyses were used to investigate the relationship between IA and resilience and their contributions to depressive mood. We then performed insula seed-based functional connectivity analyzes to identify insula networks involved in IA, resilience and depressive mood. RESULTS We first demonstrated that resilience mediates the relationship between IA and depressive mood. Second, shared and distinct intra-insula, insula-cortical and insula-subcortical networks were associated with IA, resilience and also predicted the degree of experienced depressive mood. Third, while resilience was associated with stronger insula-precuneus, insula-cerebellum and insula-prefrontal networks, IA was linked with stronger intra-insula, insula-striatum and insula-motor networks. CONCLUSIONS Our findings help understand the roles of insula-cortico-subcortical networks in IA and resilience. These results also highlight the potential use of insula networks as biomarkers for depression prediction.
Collapse
Affiliation(s)
- Alan S R Fermin
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan.
| | - Takafumi Sasaoka
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Toru Maekawa
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Kentaro Ono
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Hui-Ling Chan
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| | - Shigeto Yamawaki
- Center for Brain, Mind and Kansei Sciences Research, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
12
|
Frazer SA, Baghbanzadeh M, Rahnavard A, Crandall KA, Oakley TH. Discovering genotype-phenotype relationships with machine learning and the Visual Physiology Opsin Database (VPOD). Gigascience 2024; 13:giae073. [PMID: 39460934 PMCID: PMC11512451 DOI: 10.1093/gigascience/giae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/25/2024] [Accepted: 09/01/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Predicting phenotypes from genetic variation is foundational for fields as diverse as bioengineering and global change biology, highlighting the importance of efficient methods to predict gene functions. Linking genetic changes to phenotypic changes has been a goal of decades of experimental work, especially for some model gene families, including light-sensitive opsin proteins. Opsins can be expressed in vitro to measure light absorption parameters, including λmax-the wavelength of maximum absorbance-which strongly affects organismal phenotypes like color vision. Despite extensive research on opsins, the data remain dispersed, uncompiled, and often challenging to access, thereby precluding systematic and comprehensive analyses of the intricate relationships between genotype and phenotype. RESULTS Here, we report a newly compiled database of all heterologously expressed opsin genes with λmax phenotypes that we call the Visual Physiology Opsin Database (VPOD). VPOD_1.0 contains 864 unique opsin genotypes and corresponding λmax phenotypes collected across all animals from 73 separate publications. We use VPOD data and deepBreaks to show regression-based machine learning (ML) models often reliably predict λmax, account for nonadditive effects of mutations on function, and identify functionally critical amino acid sites. CONCLUSION The ability to reliably predict functions from gene sequences alone using ML will allow robust exploration of molecular-evolutionary patterns governing phenotype, will inform functional and evolutionary connections to an organism's ecological niche, and may be used more broadly for de novo protein design. Together, our database, phenotype predictions, and model comparisons lay the groundwork for future research applicable to families of genes with quantifiable and comparable phenotypes.
Collapse
Affiliation(s)
- Seth A Frazer
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, California 93106, USA
| | - Mahdi Baghbanzadeh
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Keith A Crandall
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
- Department of Invertebrate Zoology, National Museum of Natural History, Smithsonian Institution, Washington, DC 20012, USA
| | - Todd H Oakley
- Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, California 93106, USA
| |
Collapse
|
13
|
Faltus T, Freise J, Fluck C, Zillmann H. Ethics and regulation of neuronal optogenetics in the European Union. Pflugers Arch 2023; 475:1505-1517. [PMID: 37996706 PMCID: PMC10730653 DOI: 10.1007/s00424-023-02888-8] [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: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
Neuronal optogenetics is a technique to control the activity of neurons with light. This is achieved by artificial expression of light-sensitive ion channels in the target cells. By optogenetic methods, cells that are naturally light-insensitive can be made photosensitive and addressable by illumination and precisely controllable in time and space. So far, optogenetics has primarily been a basic research tool to better understand the brain. However, initial studies are already investigating the possibility of using optogenetics in humans for future therapeutic approaches for neuronal based diseases such as Parkinson's disease, epilepsy, or to promote stroke recovery. In addition, optogenetic methods have already been successfully applied to a human in an experimental setting. Neuronal optogenetics also raises ethical and legal issues, e.g., in relation to, animal experiments, and its application in humans. Additional ethical and legal questions may arise when optogenetic methods are investigated on cerebral organoids. Thus, for the successful translation of optogenetics from basic research to medical practice, the ethical and legal questions of this technology must also be answered, because open ethical and legal questions can hamper the translation. The paper provides an overview of the ethical and legal issues raised by neuronal optogenetics. In addition, considering the technical prerequisites for translation, the paper shows consistent approaches to address these open questions. The paper also aims to support the interdisciplinary dialogue between scientists and physicians on the one hand, and ethicists and lawyers on the other, to enable an interdisciplinary coordinated realization of neuronal optogenetics.
Collapse
Affiliation(s)
- Timo Faltus
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Johannes Freise
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Carsten Fluck
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hans Zillmann
- Law School, Faculty of Law, Economics and Business, Martin Luther University Halle-Wittenberg, Halle, Germany.
| |
Collapse
|
14
|
Hong J, Li JN, Wu FL, Bao SY, Sun HX, Zhu KH, Cai ZP, Li F, Li YQ. Projections from anteromedial thalamus nucleus to the midcingulate cortex mediate pain and anxiety-like behaviors in mice. Neurochem Int 2023; 171:105640. [PMID: 37951541 DOI: 10.1016/j.neuint.2023.105640] [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: 09/05/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Prior research has demonstrated the involvement of the midcingulate cortex (MCC) and its downstream pathway in pain regulation. However, the mechanism via which pain information is conveyed to the MCC remains unclear. The present study utilized immunohistochemistry, chemogenetics, optogenetics, and behavior detection methods to explore the involvement of MCC, anteromedial thalamus nucleus (AM), and AM-MCC pathway in pain and emotional regulation. Chemogenetics or optogenetics methods were employed to activate/inhibit MCCCaMKIIα, AMCaMKIIα, AMCaMKIIα-MCC pathway. This manipulation evokes/relieves mechanical and partial heat hyperalgesia, as well as anxiety-like behaviors. In the complete Freund,s adjuvant (CFA) inflammatory pain model, chemogenetic inhibition of the AMCaMKIIα-MCCCaMKIIα pathway contributed to pain relief. Notably, this study presented the first evidence implicating the AM in the regulation of nociception and negative emotions. Additionally, it was observed that the MCC primarily receives projections from the AM, highlighting the crucial role of this pathway in the transmission of pain and emotional information.
Collapse
Affiliation(s)
- Jie Hong
- Department of Human Anatomy, Baotou Medical College, Baotou, 014040, China; Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jia-Ni Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Feng-Ling Wu
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China; Department of Human Anatomy, College of Preclinical Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shang-Yi Bao
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China
| | - Han-Xue Sun
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China; Department of Human Anatomy, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Ke-Hua Zhu
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zhi-Ping Cai
- Department of Human Anatomy, Baotou Medical College, Baotou, 014040, China
| | - Fei Li
- Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yun-Qing Li
- Department of Human Anatomy, Baotou Medical College, Baotou, 014040, China; Department of Anatomy, Histology and Embryology and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an, 710032, China; Department of Anatomy, Basic Medical College, Dali University, Dali, 671000, China.
| |
Collapse
|
15
|
Mizuno Y, Ashok AH, Bhat BB, Jauhar S, Howes OD. Dopamine in major depressive disorder: A systematic review and meta-analysis of in vivo imaging studies. J Psychopharmacol 2023; 37:1058-1069. [PMID: 37811803 PMCID: PMC10647912 DOI: 10.1177/02698811231200881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of global disability. Several lines of evidence implicate the dopamine system in its pathophysiology. However, the magnitude and consistency of the findings are unknown. We address this by systematically reviewing in vivo imaging evidence for dopamine measures in MDD and meta-analysing these where there are sufficient studies. METHODS Studies investigating the dopaminergic system using positron emission tomography or single photon emission computed tomography in MDD and a control group were included. Demographic, clinical and imaging measures were extracted from each study, and meta-analyses and sensitivity analyses were conducted. RESULTS We identified 43 studies including 662 patients and 801 controls. Meta-analysis of 38 studies showed no difference in mean or mean variability of striatal D2/3 receptor availability (g = 0.06, p = 0.620), or combined dopamine synthesis and release capacity (g = 0.19, p = 0.309). Dopamine transporter (DAT) availability was lower in the MDD group in studies using DAT selective tracers (g = -0.56, p = 0.006), but not when tracers with an affinity for serotonin transporters were included (g = -0.21, p = 0.420). Subgroup analysis showed greater dopamine release (g = 0.49, p = 0.030), but no difference in dopamine synthesis capacity (g = -0.21, p = 0.434) in the MDD group. Striatal D1 receptor availability was lower in patients with MDD in two studies. CONCLUSIONS The meta-analysis indicates striatal DAT availability is lower, but D2/3 receptor availability is not altered in people with MDD compared to healthy controls. There may be greater dopamine release and lower striatal D1 receptors in MDD, although further studies are warranted. We discuss factors associated with these findings, discrepancies with preclinical literature and implications for future research.
Collapse
Affiliation(s)
- Yuya Mizuno
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Abhishekh Hulegar Ashok
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
- Department of Radiology, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Sameer Jauhar
- South London and Maudsley NHS Foundation Trust, London, UK
- Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Oliver D Howes
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, London, UK
- Psychiatric Imaging Group, Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
16
|
Zhang HM, Chen ZY. Electroacupuncture alleviates depression-like behaviours via a neural mechanism involving activation of Nucleus Accumbens Shell. World J Biol Psychiatry 2023; 24:721-729. [PMID: 36476217 DOI: 10.1080/15622975.2022.2155993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 12/03/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES This study investigated the effects of electroacupuncture (EA) on the depression-like behaviours in a mouse model of chronic restraint stress (CRS) and explored the underlying neural mechanisms. METHODS Depression-like behaviours including sucrose preference test (SPT), open field test (OFT) and tail suspension test (TST) were carried out to evaluate the effects of CRS and EA treatment. Using immunohistochemistry to measure the expression of c-Fos. The Nucleus Accumbens Shell (NAc Shell) in C57BL/6J mice were activated or inhibited using Chemogenetics. RESULTS All the CRS stimulated groups showed lower sucrose preference in the SPT and decreased centre times in the OFT, and increased immobility time in the TST when compared to the normal control. Interestingly, EA at LR3 or HT7 exerted anti-depressant effects, and LR3 EA exhibited a more significant restoration than HT7. Furthermore, EA at LR3 increased expression of c-Fos in the NAc Shell. Chemogenetic inhibition of NAc Shell blocked the effects of EA, whereas enhancement of NAc Shell activity profoundly reversed depressive phenotypes. CONCLUSIONS LR3 EA was effective in alleviating the depressive-like behaviours, and this therapeutic effect was associated with the activation of NAc Shell. Collectively, these findings revealed that EA may represent a promising therapeutic strategy for depression.
Collapse
Affiliation(s)
- Hua-Min Zhang
- Institute of Brain Science, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe-Yu Chen
- Institute of Brain Science, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Kook G, Jo Y, Oh C, Liang X, Kim J, Lee SM, Kim S, Choi JW, Lee HJ. Multifocal skull-compensated transcranial focused ultrasound system for neuromodulation applications based on acoustic holography. MICROSYSTEMS & NANOENGINEERING 2023; 9:45. [PMID: 37056421 PMCID: PMC10085992 DOI: 10.1038/s41378-023-00513-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/20/2023] [Accepted: 02/14/2023] [Indexed: 06/05/2023]
Abstract
Transcranial focused ultrasound stimulation is a promising therapeutic modality for human brain disorders because of its noninvasiveness, long penetration depth, and versatile spatial control capability through beamforming and beam steering. However, the skull presents a major hurdle for successful applications of ultrasound stimulation. Specifically, skull-induced focal aberration limits the capability for accurate and versatile targeting of brain subregions. In addition, there lacks a fully functional preclinical neuromodulation system suitable to conduct behavioral studies. Here, we report a miniature ultrasound system for neuromodulation applications that is capable of highly accurate multiregion targeting based on acoustic holography. Our work includes the design and implementation of an acoustic lens for targeting brain regions with compensation for skull aberration through time-reversal recording and a phase conjugation mirror. Moreover, we utilize MEMS and 3D-printing technology to implement a 0.75-g lightweight neuromodulation system and present in vivo characterization of the packaged system in freely moving mice. This preclinical system is capable of accurately targeting the desired individual or multitude of brain regions, which will enable versatile and explorative behavior studies using ultrasound neuromodulation to facilitate widespread clinical adoption.
Collapse
Affiliation(s)
- Geon Kook
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Yehhyun Jo
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Chaerin Oh
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Xiaojia Liang
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Jaewon Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Sang-Mok Lee
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Subeen Kim
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Jung-Woo Choi
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
| | - Hyunjoo Jenny Lee
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 South Korea
- KAIST Institute for NanoCentury (KINC), 291 Daehak-ro, Yuseong-gu, Daejeon, 34141 Republic of Korea
| |
Collapse
|
18
|
Wang M, Li P, Li Z, da Silva BS, Zheng W, Xiang Z, He Y, Xu T, Cordeiro C, Deng L, Dai Y, Ye M, Lin Z, Zhou J, Zhou X, Ye F, Cunha RA, Chen J, Guo W. Lateral septum adenosine A 2A receptors control stress-induced depressive-like behaviors via signaling to the hypothalamus and habenula. Nat Commun 2023; 14:1880. [PMID: 37019936 PMCID: PMC10076302 DOI: 10.1038/s41467-023-37601-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 03/23/2023] [Indexed: 04/07/2023] Open
Abstract
Major depressive disorder ranks as a major burden of disease worldwide, yet the current antidepressant medications are limited by frequent non-responsiveness and significant side effects. The lateral septum (LS) is thought to control of depression, however, the cellular and circuit substrates are largely unknown. Here, we identified a subpopulation of LS GABAergic adenosine A2A receptors (A2AR)-positive neurons mediating depressive symptoms via direct projects to the lateral habenula (LHb) and the dorsomedial hypothalamus (DMH). Activation of A2AR in the LS augmented the spiking frequency of A2AR-positive neurons leading to a decreased activation of surrounding neurons and the bi-directional manipulation of LS-A2AR activity demonstrated that LS-A2ARs are necessary and sufficient to trigger depressive phenotypes. Thus, the optogenetic modulation (stimulation or inhibition) of LS-A2AR-positive neuronal activity or LS-A2AR-positive neurons projection terminals to the LHb or DMH, phenocopied depressive behaviors. Moreover, A2AR are upregulated in the LS in two male mouse models of repeated stress-induced depression. This identification that aberrantly increased A2AR signaling in the LS is a critical upstream regulator of repeated stress-induced depressive-like behaviors provides a neurophysiological and circuit-based justification of the antidepressant potential of A2AR antagonists, prompting their clinical translation.
Collapse
Affiliation(s)
- Muran Wang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Peijun Li
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
| | - Zewen Li
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Beatriz S da Silva
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- Portuguese National Institute of Legal Medicine and Forensic Sciences (INMLCF, IP), Coimbra, Portugal
| | - Wu Zheng
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Zhenghua Xiang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology, Ministry of Education, Naval Medical University, Shanghai, China
| | - Yan He
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Tao Xu
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Cristina Cordeiro
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- Portuguese National Institute of Legal Medicine and Forensic Sciences (INMLCF, IP), Coimbra, Portugal
| | - Lu Deng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, 325000, Zhejiang Province, China
| | - Yuwei Dai
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Mengqian Ye
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Zhiqing Lin
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jianhong Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xuzhao Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Fenfen Ye
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Rodrigo A Cunha
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Jiangfan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
19
|
Increasing Adiponectin Signaling by Sub-Chronic AdipoRon Treatment Elicits Antidepressant- and Anxiolytic-Like Effects Independent of Changes in Hippocampal Plasticity. Biomedicines 2023; 11:biomedicines11020249. [PMID: 36830788 PMCID: PMC9953351 DOI: 10.3390/biomedicines11020249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
(1) Background: Adiponectin is an adipocyte-secreted hormone that has antidepressant- and anxiolytic-like effects in preclinical studies. Here, we investigated the antidepressant- and anxiolytic-like effects of sub-chronic treatment with AdipoRon, an adiponectin receptor agonist, and its potential linkage to changes in hippocampal adult neurogenesis and synaptic plasticity. (2) Methods: Different cohorts of wild-type C57BL/6J and CamKIIα-Cre male mice were treated with sub-chronic (7 days) AdipoRon, followed by behavioral, molecular, and electrophysiological experiments. (3) Results: 7-day AdipoRon treatment elicited antidepressant- and anxiolytic-like effects but did not affect hippocampal neurogenesis. AdipoRon treatment reduced hippocampal brain-derived neurotrophic factor (BDNF) levels, neuronal activation in the ventral dentate gyrus, and long-term potentiation of the perforant path. The knockdown of N-methyl-D-aspartate (NMDA) receptor subunits GluN2A and GluN2B in the ventral hippocampus did not affect the antidepressant- and anxiolytic-like effects of AdipoRon. (4) Conclusions: Increasing adiponectin signaling through sub-chronic AdipoRon treatment results in antidepressant- and anxiolytic-like effects independent of changes in hippocampal structural and synaptic function.
Collapse
|
20
|
Brown AC. Optogenetics Sheds Light on Brown and Beige Adipocytes. JOURNAL OF CELLULAR SIGNALING 2023; 4:178-186. [PMID: 37946877 PMCID: PMC10635576 DOI: 10.33696/signaling.4.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Excessive food intake leads to lipid accumulation in white adipose tissue, triggering inflammation, cellular stress, insulin resistance, and metabolic syndrome. In contrast, the dynamic energy expenditure and heat generation of brown and beige adipose tissue, driven by specialized mitochondria, render it an appealing candidate for therapeutic strategies aimed at addressing metabolic disorders. This review examines the therapeutic potential of brown and beige adipocytes for obesity and metabolic disorders, focusing on recent studies that employ optogenetics for thermogenesis control in these cells. The findings delve into the mechanisms underlying UCP1-dependent and UCP1-independent thermogenesis and how optogenetic approaches can be used to precisely modulate energy expenditure and induce thermogenesis. The convergence of adipocyte biology and optogenetics presents an exciting frontier in combating metabolic disorders and advancing our understanding of cellular regulation and energy balance.
Collapse
Affiliation(s)
- Aaron Clifford Brown
- MaineHealth Institute for Research, 81 Research Drive, Scarborough, ME 04074, USA
- School of Biomedical Sciences and Engineering, The University of Maine, Orono, Maine 04469, USA
- Tufts University School of Medicine, 145 Harrison Ave, Boston, MA 02111, USA
| |
Collapse
|
21
|
Zalachoras I, Ramos-Fernández E, Hollis F, Trovo L, Rodrigues J, Strasser A, Zanoletti O, Steiner P, Preitner N, Xin L, Astori S, Sandi C. Glutathione in the nucleus accumbens regulates motivation to exert reward-incentivized effort. eLife 2022; 11:77791. [DOI: 10.7554/elife.77791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 10/17/2022] [Indexed: 11/09/2022] Open
Abstract
Emerging evidence is implicating mitochondrial function and metabolism in the nucleus accumbens in motivated performance. However, the brain is vulnerable to excessive oxidative insults resulting from neurometabolic processes, and whether antioxidant levels in the nucleus accumbens contribute to motivated performance is not known. Here, we identify a critical role for glutathione (GSH), the most important endogenous antioxidant in the brain, in motivation. Using proton magnetic resonance spectroscopy at ultra-high field in both male humans and rodent populations, we establish that higher accumbal GSH levels are highly predictive of better, and particularly, steady performance over time in effort-related tasks. Causality was established in in vivo experiments in rats that, first, showed that downregulating GSH levels through micro-injections of the GSH synthesis inhibitor buthionine sulfoximine in the nucleus accumbens impaired effort-based reward-incentivized performance. In addition, systemic treatment with the GSH precursor N-acetyl-cysteine increased accumbal GSH levels in rats and led to improved performance, potentially mediated by a cell-type-specific shift in glutamatergic inputs to accumbal medium spiny neurons. Our data indicate a close association between accumbal GSH levels and an individual’s capacity to exert reward-incentivized effort over time. They also suggest that improvement of accumbal antioxidant function may be a feasible approach to boost motivation.
Collapse
Affiliation(s)
- Ioannis Zalachoras
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Eva Ramos-Fernández
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Fiona Hollis
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine
| | - Laura Trovo
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - João Rodrigues
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Alina Strasser
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Pascal Steiner
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - Nicolas Preitner
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé SA, Vers-chez-les-Blanc
| | - Lijing Xin
- Animal Imaging and Technology Core (AIT), Center for Biomedical Imaging (CIBM), EPFL
| | - Simone Astori
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| | - Carmen Sandi
- Laboratory of Behavioral Genetics (LGC), Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL)
| |
Collapse
|
22
|
Li H, Sung HH, Lau CG. Activation of Somatostatin-Expressing Neurons in the Lateral Septum Improves Stress-Induced Depressive-like Behaviors in Mice. Pharmaceutics 2022; 14:pharmaceutics14102253. [PMID: 36297687 PMCID: PMC9607457 DOI: 10.3390/pharmaceutics14102253] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/15/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Depression is a debilitating mood disorder with highly heterogeneous pathogenesis. The limbic system is well-linked to depression. As an important node in the limbic system, the lateral septum (LS) can modulate multiple affective and motivational behaviors. However, the role of LS in depression remains unclear. By using c-Fos expression mapping, we first screened and showed activation of the LS in various depression-related behavioral tests, including the forced swim test (FST), tail suspension test (TST), and sucrose preference test. In the LS, more than 10% of the activated neurons were somatostatin-expressing (SST) neurons. We next developed a microendoscopic calcium imaging method in freely moving mice and revealed that LSSST neural activity increased during mobility in the TST but not open field test. We hypothesize that LSSST neuronal activity is linked to stress and depression. In two mouse models of depression, repeated lipopolysaccharide (LPS) injection and chronic restraint stress (CRS), we showed that LS neuronal activation was suppressed. To examine whether the re-activation of LSSST neurons can be therapeutically beneficial, we optogenetically activated LSSST neurons and produced antidepressant-like effects in LPS-injected mice by increasing TST motility. Moreover, chemogenetic activation of LSSST neurons increased FST struggling in the CRS-exposed mice. Together, these results provide the first evidence of a role for LSSST neurons in regulating depressive-like behaviors in mice and identify them as a potential therapeutic target for neuromodulation-based intervention in depression.
Collapse
Affiliation(s)
- Huanhuan Li
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Hyun Hailey Sung
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| | - Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Hong Kong 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
- Correspondence: ; Tel.: +852-3442-4345
| |
Collapse
|
23
|
Uliana DL, Zhu X, Gomes FV, Grace AA. Using animal models for the studies of schizophrenia and depression: The value of translational models for treatment and prevention. Front Behav Neurosci 2022; 16:935320. [PMID: 36090659 PMCID: PMC9449416 DOI: 10.3389/fnbeh.2022.935320] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/04/2022] [Indexed: 11/29/2022] Open
Abstract
Animal models of psychiatric disorders have been highly effective in advancing the field, identifying circuits related to pathophysiology, and identifying novel therapeutic targets. In this review, we show how animal models, particularly those based on development, have provided essential information regarding circuits involved in disorders, disease progression, and novel targets for intervention and potentially prevention. Nonetheless, in recent years there has been a pushback, largely driven by the US National Institute of Mental Health (NIMH), to shift away from animal models and instead focus on circuits in normal subjects. This has been driven primarily from a lack of discovery of new effective therapeutic targets, and the failure of targets based on preclinical research to show efficacy. We discuss why animal models of complex disorders, when strongly cross-validated by clinical research, are essential to understand disease etiology as well as pathophysiology, and direct new drug discovery. Issues related to shortcomings in clinical trial design that confound translation from animal models as well as the failure to take patient pharmacological history into account are proposed to be a source of the failure of what are likely effective compounds from showing promise in clinical trials.
Collapse
Affiliation(s)
- Daniela L. Uliana
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Xiyu Zhu
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Felipe V. Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A. Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
24
|
Elevated BICD2 DNA methylation in blood of major depressive disorder patients and reduction of depressive-like behaviors in hippocampal Bicd2-knockdown mice. Proc Natl Acad Sci U S A 2022; 119:e2201967119. [PMID: 35858435 PMCID: PMC9335189 DOI: 10.1073/pnas.2201967119] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Major depressive disorder (MDD) is a prevalent and devastating mental illness. To date, the diagnosis of MDD is largely dependent on clinical interviews and questionnaires and still lacks a reliable biomarker. DNA methylation has a stable and reversible nature and is likely associated with the course and therapeutic efficacy of complex diseases, which may play an important role in the etiology of a disease. Here, we identified and validated a DNA methylation biomarker for MDD from four independent cohorts of the Chinese Han population. First, we integrated the analysis of the DNA methylation microarray (n = 80) and RNA expression microarray data (n = 40) and identified BICD2 as the top-ranked gene. In the replication phase, we employed the Sequenom MassARRAY method to confirm the DNA hypermethylation change in a large sample size (n = 1,346) and used the methylation-sensitive restriction enzymes and a quantitative PCR approach (MSE-qPCR) and qPCR method to confirm the correlation between DNA hypermethylation and mRNA down-regulation of BICD2 (n = 60). The results were replicated in the peripheral blood of mice with depressive-like behaviors, while in the hippocampus of mice, Bicd2 showed DNA hypomethylation and mRNA/protein up-regulation. Hippocampal Bicd2 knockdown demonstrates antidepressant action in the chronic unpredictable mild stress (CUMS) mouse model of depression, which may be mediated by increased BDNF expression. Our study identified a potential DNA methylation biomarker and investigated its functional implications, which could be exploited to improve the diagnosis and treatment of MDD.
Collapse
|
25
|
Kim J, Kang S, Choi TY, Chang KA, Koo JW. Metabotropic Glutamate Receptor 5 in Amygdala Target Neurons Regulates Susceptibility to Chronic Social Stress. Biol Psychiatry 2022; 92:104-115. [PMID: 35314057 DOI: 10.1016/j.biopsych.2022.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGluR5) has been implicated in stress-related psychiatric disorders, particularly major depressive disorder. Although growing evidence supports the proresilient role of mGluR5 in corticolimbic circuitry in the depressive-like behaviors following chronic stress exposure, the underlying neural mechanisms, including circuits and molecules, remain unknown. METHODS We measured the c-Fos expression and probability of neurotransmitter release in and from basolateral amygdala (BLA) neurons projecting to the medial prefrontal cortex (mPFC) and to the ventral hippocampus (vHPC) after chronic social defeat stress. The role of BLA projections in depressive-like behaviors was assessed using optogenetic manipulations, and the underlying molecular mechanisms of mGluR5 and downstream signaling were investigated by Western blotting, viral-mediated gene transfer, and pharmacological manipulations. RESULTS Chronic social defeat stress disrupted neural activity and glutamatergic transmission in both BLA projections. Optogenetic activation of BLA projections reversed the detrimental effects of chronic social defeat stress on depressive-like behaviors and mGluR5 expression in the mPFC and vHPC. Conversely, inhibition of BLA projections of mice undergoing subthreshold social defeat stress induced a susceptible phenotype and mGluR5 reduction. These two BLA circuits appeared to act in an independent way. We demonstrate that mGluR5 overexpression in the mPFC or vHPC was proresilient while the mGluR5 knockdown was prosusceptible and that the proresilient effects of mGluR5 are mediated through distinctive downstream signaling pathways in the mPFC and vHPC. CONCLUSIONS These findings identify mGluR5 in the mPFC and vHPC that receive BLA inputs as a critical mediator of stress resilience, highlighting circuit-specific signaling for depressive-like behaviors.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
26
|
Xu S, Yao X, Li B, Cui R, Zhu C, Wang Y, Yang W. Uncovering the Underlying Mechanisms of Ketamine as a Novel Antidepressant. Front Pharmacol 2022; 12:740996. [PMID: 35872836 PMCID: PMC9301111 DOI: 10.3389/fphar.2021.740996] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) is a devastating psychiatric disorder which exacts enormous personal and social-economic burdens. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has been discovered to exert rapid and sustained antidepressant-like actions on MDD patients and animal models. However, the dissociation and psychotomimetic propensities of ketamine have limited its use for psychiatric indications. Here, we review recently proposed mechanistic hypotheses regarding how ketamine exerts antidepressant-like actions. Ketamine may potentiate α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR)-mediated transmission in pyramidal neurons by disinhibition and/or blockade of spontaneous NMDAR-mediated neurotransmission. Ketamine may also activate neuroplasticity- and synaptogenesis-relevant signaling pathways, which may converge on key components like brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) and mechanistic target of rapamycin (mTOR). These processes may subsequently rebalance the excitatory/inhibitory transmission and restore neural network integrity that is compromised in depression. Understanding the mechanisms underpinning ketamine’s antidepressant-like actions at cellular and neural circuit level will drive the development of safe and effective pharmacological interventions for the treatment of MDD.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Yao Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| |
Collapse
|
27
|
Uliana DL, Gomes FV, Grace AA. Nucleus reuniens inactivation reverses stress-induced hypodopaminergic state and altered hippocampal-accumbens synaptic plasticity. Neuropsychopharmacology 2022; 47:1513-1522. [PMID: 35488085 PMCID: PMC9205859 DOI: 10.1038/s41386-022-01333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 03/11/2022] [Accepted: 04/19/2022] [Indexed: 11/09/2022]
Abstract
The nucleus reuniens of the thalamus (RE) is a pivotal area responsible for the connectivity of the prefrontal-hippocampus pathway that regulates cognitive, executive, and fear learning processes. Recently, it was proposed that the RE participates in the pathophysiological states related to affective dysregulation. We investigated the role of RE in motivational behavioral and electrophysiological dysregulation induced by stress. Adult Sprague-Dawley rats were exposed to a combination of stressors (restraint stress+footshock) for 10 days and tested one to two weeks later in the forced swim test (FST), ventral tegmental area (VTA)dopamine (DA) neuron electrophysiological activity, and hippocampal-nucleus accumbens plasticity. The RE was inactivated by injecting TTX prior to the procedures. The stress exposure increased the immobility in the FST and decreased VTA DA neuron population activity. Whereas an early long-term potentiation (e-LTP) in the ventral hippocampus-nucleus accumbens pathway was found after fimbria high-frequency stimulation in naïve animals, stressed animals showed an early long-term depression (e-LTD). Inactivation of the RE reversed the stress-induced changes in the FST and restored dopaminergic activity. RE inactivation partially recovered the stress-induced abnormal hippocampal-accumbens plasticity observed in controls. Our findings support the role of the RE in regulating affective dysregulation and blunted VTA DA system function induced by stress. Also, it points to the hippocampal-accumbens pathway as a potential neural circuit through which RE could modulate activity. Therefore, RE may represent a key brain region involved in the neurobiology of amotivational states and may provide insights into circuit dysfunction and markers of the maladaptive stress response.
Collapse
Affiliation(s)
- Daniela L. Uliana
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA
| | - Felipe V. Gomes
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA ,grid.11899.380000 0004 1937 0722Present Address: Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Anthony A. Grace
- grid.21925.3d0000 0004 1936 9000Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA USA
| |
Collapse
|
28
|
Brondi M, Bruzzone M, Lodovichi C, dal Maschio M. Optogenetic Methods to Investigate Brain Alterations in Preclinical Models. Cells 2022; 11:1848. [PMID: 35681542 PMCID: PMC9180859 DOI: 10.3390/cells11111848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 02/05/2023] Open
Abstract
Investigating the neuronal dynamics supporting brain functions and understanding how the alterations in these mechanisms result in pathological conditions represents a fundamental challenge. Preclinical research on model organisms allows for a multiscale and multiparametric analysis in vivo of the neuronal mechanisms and holds the potential for better linking the symptoms of a neurological disorder to the underlying cellular and circuit alterations, eventually leading to the identification of therapeutic/rescue strategies. In recent years, brain research in model organisms has taken advantage, along with other techniques, of the development and continuous refinement of methods that use light and optical approaches to reconstruct the activity of brain circuits at the cellular and system levels, and to probe the impact of the different neuronal components in the observed dynamics. These tools, combining low-invasiveness of optical approaches with the power of genetic engineering, are currently revolutionizing the way, the scale and the perspective of investigating brain diseases. The aim of this review is to describe how brain functions can be investigated with optical approaches currently available and to illustrate how these techniques have been adopted to study pathological alterations of brain physiology.
Collapse
Affiliation(s)
- Marco Brondi
- Institute of Neuroscience, National Research Council-CNR, Viale G. Colombo 3, 35121 Padova, Italy; (M.B.); (C.L.)
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy
| | - Matteo Bruzzone
- Department of Biomedical Sciences, Università degli Studi di Padova, Via U. Bassi 58B, 35121 Padova, Italy;
- Padova Neuroscience Center (PNC), Università degli Studi di Padova, Via Orus 2, 35129 Padova, Italy
| | - Claudia Lodovichi
- Institute of Neuroscience, National Research Council-CNR, Viale G. Colombo 3, 35121 Padova, Italy; (M.B.); (C.L.)
- Veneto Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy
- Department of Biomedical Sciences, Università degli Studi di Padova, Via U. Bassi 58B, 35121 Padova, Italy;
- Padova Neuroscience Center (PNC), Università degli Studi di Padova, Via Orus 2, 35129 Padova, Italy
| | - Marco dal Maschio
- Department of Biomedical Sciences, Università degli Studi di Padova, Via U. Bassi 58B, 35121 Padova, Italy;
- Padova Neuroscience Center (PNC), Università degli Studi di Padova, Via Orus 2, 35129 Padova, Italy
| |
Collapse
|
29
|
Tan Z, Yang G, Qiu J, Yan W, Liu Y, Ma Z, Li J, Liu J, Shan N. Quercetin Alleviates Demyelination Through Regulating Microglial Phenotype Transformation to Mitigate Neuropsychiatric Symptoms in Mice with Vascular Dementia. Mol Neurobiol 2022; 59:3140-3158. [PMID: 35267135 DOI: 10.1007/s12035-021-02712-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
Abstract
Cerebral hypoperfusion plays a pivotal role in the ictus and development of vascular dementia (VaD) with neuropsychiatric symptoms. To date, few pharmacological interventions for neuropsychiatric symptoms are available in the VaD patients with neuropsychiatric impairments. Here, our results demonstrated that the extent of demyelination was dramatically deteriorated and the thickness of myelin sheath was evidently decreased in the presence of cerebral hypoperfusion, whereas Quercetin possessed the potential of abrogating these effects at least in part, then relieving anxiety and depression-like behavior when mice exposed to bilateral carotid artery stenosis (BCAS)/chronic restraint stress (CRS). The underlying mechanism was that Quercetin facilitated secretion of anti-inflammatory cytokines (IL-4 and IL-10) and in turn decreased production of pro-inflammatory factors (TNF-α and IL-1β) due to regulating microglial phenotype transformation, thereafter enhancing the microglial engulfment ability of myelin fragments in vitro and in vivo. Collectively, the results demonstrated that that Quercetin mediated microglial transformation into anti-inflammatory phenotype to reduce demyelination in ventral hippocampus (vHIP), thereafter mitigating neuropsychiatric deficits (including anxiety and depression). The present research broadens the therapeutic scope of Quercetin in central nervous system (CNS) disorders with presence of white matter damage and/or the insufficient activation of anti-inflammatory microglia, particularly for vascular dementia with/without neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Zihu Tan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Guang Yang
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Jing Qiu
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Wenjing Yan
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Yu Liu
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China
| | - Zhengling Ma
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, 430061, China
| | - Jia Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment By Acupuncture and Moxibustion, Wuhan, 430061, China
| | - Jing Liu
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment By Acupuncture and Moxibustion, Wuhan, 430061, China
| | - Nan Shan
- Department of Geriatrics, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China. .,Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China. .,Hubei Provincial Academy of Traditional Chinese Medicine, 430061, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
30
|
Sex Differences in Anxiety and Depression: What Can (and Cannot) Preclinical Studies Tell Us? SEXES 2022. [DOI: 10.3390/sexes3010012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In recent years, the gender perspective in scientific research and sex differences in biological studies on emotional disorders have become increasingly important. However, sex bias in basic research on anxiety and depression is still far from being covered. This review addresses the study of sex differences in the field of anxiety and depression using animal models that consider this issue so far. What can preclinical studies tell us and what are their main limitations? First, we describe the behavioral tests most frequently used in preclinical research to assess depressive-like and anxiety-like behaviors in rodents. Then, we analyze the main findings, strengths, and weaknesses of rodent models of anxiety and depression, dividing them into three main categories: sex chromosome complement-biased sex differences; gonadal hormone-biased sex differences; environmental-biased sex differences. Regardless of the animal model used, none can reproduce all the characteristics of such complex and multifactorial pathologies as anxiety and depressive disorders; however, each animal model contributes to elucidating the bases that underlie these disorders. The importance is highlighted of considering sex differences in the responses that emerge from each model.
Collapse
|
31
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
32
|
Chen Y, Zheng Y, Yan J, Zhu C, Zeng X, Zheng S, Li W, Yao L, Xia Y, Su WW, Chen Y. Early Life Stress Induces Different Behaviors in Adolescence and Adulthood May Related With Abnormal Medial Prefrontal Cortex Excitation/Inhibition Balance. Front Neurosci 2022; 15:720286. [PMID: 35058738 PMCID: PMC8765554 DOI: 10.3389/fnins.2021.720286] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/02/2021] [Indexed: 12/28/2022] Open
Abstract
Early life stress is thought to be a risk factor for emotional disorders, particularly depression and anxiety. Although the excitation/inhibition (E/I) imbalance has been implicated in neuropsychiatric disorders, whether early life stress affects the E/I balance in the medial prefrontal cortex at various developmental stages is unclear. In this study, rats exposed to maternal separation (MS) that exhibited a well-established early life stress paradigm were used to evaluate the E/I balance in adolescence (postnatal day P43-60) and adulthood (P82-100) by behavior tests, whole-cell recordings, and microdialysis coupled with high performance liquid chromatography-mass spectrometry (HPLC-MS) analysis. First, the behavioral tests revealed that MS induced both anxiety- and depressive-like behaviors in adolescent rats but only depressive-like behavior in adult rats. Second, MS increased the action potential frequency and E/I balance of synaptic transmission onto L5 pyramidal neurons in the prelimbic (PrL) brain region of adolescent rats while decreasing the action potential frequency and E/I balance in adult rats. Finally, MS increases extracellular glutamate levels and decreased the paired-pulse ratio of evoked excitatory postsynaptic currents (EPSCs) of pyramidal neurons in the PrL of adolescent rats. In contrast, MS decreased extracellular glutamate levels and increased the paired-pulse ratio of evoked EPSCs of pyramidal neurons in the PrL of adult rats. The present results reveal a key role of E/I balance in different MS-induced disorders may related to the altered probability of presynaptic glutamate release at different developmental stages.
Collapse
Affiliation(s)
- Yiwen Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanjia Zheng
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinglan Yan
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuanan Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Xiamen Xianyue Hospital, Xiamen, China
| | - Xuan Zeng
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shaoyi Zheng
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenwen Li
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yao
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yucen Xia
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei-Wei Su
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.,Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, China.,Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China.,Guangdong Province Key Laboratory of Psychiatric Disorders, Southern Medical University, Guangzhou, China
| |
Collapse
|
33
|
Mantas I, Saarinen M, Xu ZQD, Svenningsson P. Update on GPCR-based targets for the development of novel antidepressants. Mol Psychiatry 2022; 27:534-558. [PMID: 33589739 PMCID: PMC8960420 DOI: 10.1038/s41380-021-01040-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/31/2023]
Abstract
Traditional antidepressants largely interfere with monoaminergic transport or degradation systems, taking several weeks to have their therapeutic actions. Moreover, a large proportion of depressed patients are resistant to these therapies. Several atypical antidepressants have been developed which interact with G protein coupled receptors (GPCRs) instead, as direct targeting of receptors may achieve more efficacious and faster antidepressant actions. The focus of this review is to provide an update on how distinct GPCRs mediate antidepressant actions and discuss recent insights into how GPCRs regulate the pathophysiology of Major Depressive Disorder (MDD). We also discuss the therapeutic potential of novel GPCR targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles. Finally, we highlight recent advances in understanding GPCR pharmacology and structure, and how they may provide new avenues for drug development.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Marcus Saarinen
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
34
|
Kang S, Jun S, Baek SJ, Park H, Yamamoto Y, Tanaka-Yamamoto K. Recent Advances in the Understanding of Specific Efferent Pathways Emerging From the Cerebellum. Front Neuroanat 2021; 15:759948. [PMID: 34975418 PMCID: PMC8716603 DOI: 10.3389/fnana.2021.759948] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
The cerebellum has a long history in terms of research on its network structures and motor functions, yet our understanding of them has further advanced in recent years owing to technical developments, such as viral tracers, optogenetic and chemogenetic manipulation, and single cell gene expression analyses. Specifically, it is now widely accepted that the cerebellum is also involved in non-motor functions, such as cognitive and psychological functions, mainly from studies that have clarified neuronal pathways from the cerebellum to other brain regions that are relevant to these functions. The techniques to manipulate specific neuronal pathways were effectively utilized to demonstrate the involvement of the cerebellum and its pathways in specific brain functions, without altering motor activity. In particular, the cerebellar efferent pathways that have recently gained attention are not only monosynaptic connections to other brain regions, including the periaqueductal gray and ventral tegmental area, but also polysynaptic connections to other brain regions, including the non-primary motor cortex and hippocampus. Besides these efferent pathways associated with non-motor functions, recent studies using sophisticated experimental techniques further characterized the historically studied efferent pathways that are primarily associated with motor functions. Nevertheless, to our knowledge, there are no articles that comprehensively describe various cerebellar efferent pathways, although there are many interesting review articles focusing on specific functions or pathways. Here, we summarize the recent findings on neuronal networks projecting from the cerebellum to several brain regions. We also introduce various techniques that have enabled us to advance our understanding of the cerebellar efferent pathways, and further discuss possible directions for future research regarding these efferent pathways and their functions.
Collapse
Affiliation(s)
- Seulgi Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Soo Ji Baek
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| |
Collapse
|
35
|
Dimén D, Puska G, Szendi V, Sipos E, Zelena D, Dobolyi Á. Sex-specific parenting and depression evoked by preoptic inhibitory neurons. iScience 2021; 24:103090. [PMID: 34604722 PMCID: PMC8463871 DOI: 10.1016/j.isci.2021.103090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/20/2021] [Accepted: 09/01/2021] [Indexed: 01/08/2023] Open
Abstract
The role of preoptic GABAergic inhibitory neurons was addressed in parenting, anxiety and depression. Pup exposure and forced swimming resulted in similar c-Fos activation pattern in neurons expressing vesicular GABA transporter in the preoptic area with generally stronger labeling and different distributional pattern in females than in males. Chemogenetic stimulation of preoptic GABAergic cells resulted in elevated maternal motivation and caring behavior in females and mothers but aggression toward pups in males. Behavioral effects were the opposite following inhibition of preoptic GABAergic neurons suggesting their physiological relevance. In addition, increased anxiety-like and depression-like behaviors were found following chemogenetic stimulation of the same neurons in females, whereas previous pup exposure increased only anxiety-like behavior suggesting that not the pups, but overstimulation of the cells can lead to depression-like behavior. A sexually dimorphic projection pattern of preoptic GABAergic neurons was also identified, which could mediate sex-dependent parenting and associated emotional behaviors. Preoptic GABAergic neurons promote maternal behaviors in females mice Activation of preoptic GABAergic neurons induces pup-directed aggression in males Projection pattern of preoptic GABAergic neurons is sexually dimorphic Depression-like behaviors are provoked by stimulation of preoptic GABAergic neurons
Collapse
Affiliation(s)
- Diána Dimén
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Gina Puska
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary.,Department of Ecology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Vivien Szendi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Eszter Sipos
- Department of Behavioral and Stress Studies, Institute of Experimental Medicine, 1080 Budapest, Hungary
| | - Dóra Zelena
- Department of Behavioral and Stress Studies, Institute of Experimental Medicine, 1080 Budapest, Hungary.,Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Árpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
36
|
Ben Haim L, Escartin C. Astrocytes and neuropsychiatric symptoms in neurodegenerative diseases: Exploring the missing links. Curr Opin Neurobiol 2021; 72:63-71. [PMID: 34628361 DOI: 10.1016/j.conb.2021.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 11/28/2022]
Abstract
Neurodegenerative diseases (NDs) are characterized by primary symptoms, such as cognitive or motor deficits. In addition, the presence of neuropsychiatric symptoms (NPS) in patients with ND is being increasingly acknowledged as an important disease feature. Yet, their neurobiological basis remains unclear and mostly centered on neurons while overlooking astrocytes, which are crucial regulators of neuronal function underlying complex behaviors. In this opinion article, we briefly review evidence for NPS in ND and discuss their experimental assessment in preclinical models. We then present recent studies showing that astrocyte-specific dysfunctions can lead to NPS. Because many astrocyte alterations are also observed in ND, we suggest that they might underlie ND-associated NPS. We argue that there is a need for dedicated preclinical studies assessing astrocyte-based therapeutic strategies targeting NPS in the context of ND.
Collapse
Affiliation(s)
- Lucile Ben Haim
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, 92265, Fontenay-aux-Roses, France.
| |
Collapse
|
37
|
Wang S, Leri F, Rizvi SJ. Anhedonia as a central factor in depression: Neural mechanisms revealed from preclinical to clinical evidence. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110289. [PMID: 33631251 DOI: 10.1016/j.pnpbp.2021.110289] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/25/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
Anhedonia is one of the core symptoms of major depressive disorder (MDD), which is often inadequately treated by traditional antidepressants. The modern framework of anhedonia extends the definition from impaired consummatory pleasure or interest in rewards to a broad spectrum of deficits that impact functions such as reward anticipation, approach motivation, effort expenditure, reward valuation, expectation, and reward-cue association learning. Substantial preclinical and clinical research has explored the neural basis of reward deficits in the context of depression, and has implicated mesocorticolimbic reward circuitry comprising the nucleus accumbens, ventral pallidum, ventral tegmental area, amygdala, hippocampus, anterior cingulate, insula, orbitofrontal cortex, and other prefrontal cortex regions. Dopamine modulates several reward facets including anticipation, motivation, effort, and learning. As well, serotonin, norepinephrine, opioids, glutamate, Gamma aminobutyric acid (GABA), and acetylcholine are also involved in anhedonia, and medications targeting these systems may also potentially normalize reward processing in depression. Unfortunately, whereas reward anticipation and reward outcome are extensively explored by both preclinical and clinical studies, translational gaps remain in reward motivation, effort, valuation, and learning, where clinical neuroimaging studies are in the early stages. This review aims to synthesize the neurobiological mechanisms underlying anhedonia in MDD uncovered by preclinical and clinical research. The translational difficulties in studying the neural basis of reward are also discussed.
Collapse
Affiliation(s)
- Shijing Wang
- Arthur Sommer Rotenberg Suicide and Depression Studies Program, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Francesco Leri
- Department of Psychology, University of Guelph, Ontario, Canada
| | - Sakina J Rizvi
- Arthur Sommer Rotenberg Suicide and Depression Studies Program, St. Michael's Hospital, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
38
|
Over-representation of fundamental decision variables in the prefrontal cortex underlies decision bias. Neurosci Res 2021; 173:1-13. [PMID: 34274406 DOI: 10.1016/j.neures.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/15/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022]
Abstract
The brain is organized into anatomically distinct structures consisting of a variety of projection neurons. While such evolutionarily conserved neural circuit organization underlies the innate ability of animals to swiftly adapt to environments, they can cause biased cognition and behavior. Although recent studies have begun to address the causal importance of projection-neuron types as distinct computational units, it remains unclear how projection types are functionally organized in encoding variables during cognitive tasks. This review focuses on the neural computation of decision making in the prefrontal cortex and discusses what decision variables are encoded by single neurons, neuronal populations, and projection type, alongside how specific projection types constrain decision making. We focus particularly on "over-representations" of distinct decision variables in the prefrontal cortex that reflect the biological and subjective significance of the variables for the decision makers. We suggest that task-specific over-representation in the prefrontal cortex involves the refinement of the given decision making, while generalized over-representation of fundamental decision variables is associated with suboptimal decision biases, including pathological ones such as those in patients with psychiatric disorders. Such over-representation of the fundamental decision variables in the prefrontal cortex appear to be tightly constrained by afferent and efferent connections that can be optogenetically intervened on. These ideas may provide critical insights into potential therapeutic targets for psychiatric disorders, including addiction and depression.
Collapse
|
39
|
Ivanets NN, Svistunov AA, Chubarev VN, Kinkulkina MA, Tikhonova YG, Syzrantsev NS, Sologova SS, Ignatyeva NV, Mutig K, Tarasov VV. Can Molecular Biology Propose Reliable Biomarkers for Diagnosing Major Depression? Curr Pharm Des 2021; 27:305-318. [PMID: 33234092 DOI: 10.2174/1381612826666201124110437] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 08/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Modern medicine has provided considerable knowledge of the pathophysiology of mental disorders at the body, systemic, organ and neurochemical levels of the biological organization of the body. Modern clinical diagnostics of depression have some problems, that is why psychiatric society makes use of diagnostics and taxonomy of different types of depression by implemention of modern molecular biomarkers in diagnostic procedures. But up to now, there are no reliable biomarkers of major depressive disorder (MDD) and other types of depression. OBJECTIVE The purpose of this review is to find fundamentals in pathological mechanisms of depression, which could be a basis for development of molecular and genetic biomarkers, being the most feasible for clinical use. METHOD This review summarizes the published data using PubMed, Science Direct, Google Scholar and Scopus. RESULTS In this review, we summarized and discussed findings in molecular biology, genetics, neuroplasticity, neurotransmitters, and neuroimaging that could increase our understanding of the biological foundations of depression and show new directions for the development of reliable biomarkers. We did not find any molecular and genetic biomarker approved for the clinic. But the Genome-Wide Association Study method promises some progress in the development of biomarkers based on SNP in the future. Epigenetic factors also are a promising target for biomarkers. We have found some differences in the etiology of different types of atypical and melancholic depression. This knowledge could be the basis for development of biomarkers for clinical practice in diagnosis, prognosis and selection of treatment. CONCLUSION Depression is not a monoetiological disease. Many pathological mechanisms are involved in depression, thus up to now, there is no approved and reliable biomarker for diagnosis, prognosis and correction of treatment of depression. The structural and functional complexity of the brain, the lack of invasive technology, poor correlations between genetic and clinical manifestation of depression, imperfect psychiatric classification and taxonomy of subtypes of disease are the main causes of this situation. One of the possible ways to come over this situation can be to pay attention to the trigger mechanism of disease and its subtypes. Researchers and clinicians should focus their efforts on searching the trigger mechanism of depression and different types of it . HPA axis can be a candidate for such trigger in depression caused by stress, because it influences the main branches of disease: neuroinflammation, activity of biogenic amines, oxidative and nitrosative stress, epigenetic factors, metabolomics, etc. But before we shall find any trigger mechanism, we need to create complex biomarkers reflecting genetic, epigenetic, metabolomics and other pathological changes in different types of depression. Recently the most encouraging results have been obtained from genetics and neuroimaging. Continuing research in these areas should be forced by using computational, statistical and systems biology approaches, which can allow to obtain more knowledge about the neurobiology of depression. In order to obtain clinically useful tests, search for biomarkers should use appropriate research methodologies with increasing samples and identifying more homogeneous groups of depressed patients.
Collapse
Affiliation(s)
- Nikolay N Ivanets
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Andrey A Svistunov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Vladimir N Chubarev
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Marina A Kinkulkina
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Yuliya G Tikhonova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Nikita S Syzrantsev
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Susanna S Sologova
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Nelly V Ignatyeva
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Kerim Mutig
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| | - Vadim V Tarasov
- Department of Pharmacology, Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russian Federation
| |
Collapse
|
40
|
Leite Dantas R, Freff J, Ambrée O, Beins EC, Forstner AJ, Dannlowski U, Baune BT, Scheu S, Alferink J. Dendritic Cells: Neglected Modulators of Peripheral Immune Responses and Neuroinflammation in Mood Disorders? Cells 2021; 10:941. [PMID: 33921690 PMCID: PMC8072712 DOI: 10.3390/cells10040941] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/25/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Affective disorders (AD) including major depressive disorder (MDD) and bipolar disorder (BD) are common mood disorders associated with increased disability and poor health outcomes. Altered immune responses characterized by increased serum levels of pro-inflammatory cytokines and neuroinflammation are common findings in patients with AD and in corresponding animal models. Dendritic cells (DCs) represent a heterogeneous population of myeloid cells that orchestrate innate and adaptive immune responses and self-tolerance. Upon sensing exogenous and endogenous danger signals, mature DCs secrete proinflammatory factors, acquire migratory and antigen presenting capacities and thus contribute to neuroinflammation in trauma, autoimmunity, and neurodegenerative diseases. However, little is known about the involvement of DCs in the pathogenesis of AD. In this review, we summarize the current knowledge on DCs in peripheral immune responses and neuroinflammation in MDD and BD. In addition, we consider the impact of DCs on neuroinflammation and behavior in animal models of AD. Finally, we will discuss therapeutic perspectives targeting DCs and their effector molecules in mood disorders.
Collapse
Affiliation(s)
- Rafael Leite Dantas
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Jana Freff
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| | - Oliver Ambrée
- Department of Behavioural Biology, University of Osnabrück, 49076 Osnabrück, Germany;
- Center of Cellular Nanoanalytics, University of Osnabrück, 49076 Osnabrück, Germany
| | - Eva C. Beins
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
| | - Andreas J. Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, 53127 Bonn, Germany; (E.C.B.); (A.J.F.)
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, 52428 Jülich, Germany
| | - Udo Dannlowski
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
| | - Bernhard T. Baune
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Parkville, VIC 3010, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany;
| | - Judith Alferink
- Department of Mental Health, University of Münster, 48149 Münster, Germany; (R.L.D.); (J.F.); (U.D.); (B.T.B.)
- Cells in Motion Interfaculty Centre, University of Münster, 48149 Münster, Germany
| |
Collapse
|
41
|
Lv QY, Chen MM, Li Y, Yu Y, Liao H. Brain circuit dysfunction in specific symptoms of depression. Eur J Neurosci 2021; 55:2393-2403. [PMID: 33818849 DOI: 10.1111/ejn.15221] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022]
Abstract
Since the depressive disorder manifests complex and diverse symptoms clinically, its pathological mechanism and therapeutic options are difficult to determine. In recent years, the advent of optogenetics, chemogenetics and viral tracing techniques, along with the well-established rodent model of depression, has led to a shift in the focus of depression research from single molecules to neural circuits. In virtue of the powerful tools above, psychiatric disorder such as depression could be well related to the disfunction of brain's connection. Moreover, compelling studies also support that the diversity of depressive behaviour could be involved with the discrete changes in a distinct circuit of the brain. Therefore, summarising the differential changes of the neural circuits in mice with depression-like behaviour may provide a better understanding of the causal relationships between neural circuit and depressive behaviour. Here, we focus on the changes in the neural circuitry underlying various depression-like phenotypes, including motivation, despair, social avoidance and comorbid sequelae, which may provide an explanation to circuit-specific discrepancy in depression-like behaviour.
Collapse
Affiliation(s)
- Qun Y Lv
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Ming M Chen
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Yu Li
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Yang Yu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Hong Liao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
42
|
Salmina AB, Gorina YV, Erofeev AI, Balaban PM, Bezprozvanny IB, Vlasova OL. Optogenetic and chemogenetic modulation of astroglial secretory phenotype. Rev Neurosci 2021; 32:459-479. [PMID: 33550788 DOI: 10.1515/revneuro-2020-0119] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/28/2020] [Indexed: 12/20/2022]
Abstract
Astrocytes play a major role in brain function and alterations in astrocyte function that contribute to the pathogenesis of many brain disorders. The astrocytes are attractive cellular targets for neuroprotection and brain tissue regeneration. Development of novel approaches to monitor and to control astroglial function is of great importance for further progress in basic neurobiology and in clinical neurology, as well as psychiatry. Recently developed advanced optogenetic and chemogenetic techniques enable precise stimulation of astrocytes in vitro and in vivo, which can be achieved by the expression of light-sensitive channels and receptors, or by expression of receptors exclusively activated by designer drugs. Optogenetic stimulation of astrocytes leads to dramatic changes in intracellular calcium concentrations and causes the release of gliotransmitters. Optogenetic and chemogenetic protocols for astrocyte activation aid in extracting novel information regarding the function of brain's neurovascular unit. This review summarizes current data obtained by this approach and discusses a potential mechanistic connection between astrocyte stimulation and changes in brain physiology.
Collapse
Affiliation(s)
- Alla B Salmina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Yana V Gorina
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Research Institute of Molecular Medicine and Pathobiochemistry, Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
| | - Alexander I Erofeev
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Pavel M Balaban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Ilya B Bezprozvanny
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Olga L Vlasova
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| |
Collapse
|
43
|
Coley AA, Padilla-Coreano N, Patel R, Tye KM. Valence processing in the PFC: Reconciling circuit-level and systems-level views. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 158:171-212. [PMID: 33785145 DOI: 10.1016/bs.irn.2020.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An essential component in animal behavior is the ability to process emotion and dissociate among positive and negative valence in response to a rewarding or aversive stimulus. The medial prefrontal cortex (mPFC)-responsible for higher order executive functions that include cognition, learning, and working memory; and is also involved in sociability-plays a major role in emotional processing and control. Although the amygdala is widely regarded as the "emotional hub," the mPFC encodes for context-specific salience and elicits top-down control over limbic circuitry. The mPFC can then conduct behavioral responses, via cortico-striatal and cortico-brainstem pathways, that correspond to emotional stimuli. Evidence shows that abnormalities within the mPFC lead to sociability deficits, working memory impairments, and drug-seeking behavior that include addiction and compulsive disorders; as well as conditions such as anhedonia. Recent studies investigate the effects of aberrant salience processing on cortical circuitry and neuronal populations associated with these behaviors. In this chapter, we discuss mPFC valence processing, neuroanatomical connections, and physiological substrates involved in mPFC-associated behavior. We review neurocomputational and theoretical models such as "mixed selectivity," that describe cognitive control, attentiveness, and motivational drives. Using this knowledge, we describe the effects of valence imbalances and its influence on mPFC neural pathways that contribute to deficits in social cognition, while understanding the effects in addiction/compulsive behaviors and anhedonia.
Collapse
Affiliation(s)
- Austin A Coley
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Reesha Patel
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Kay M Tye
- Salk Institute for Biological Studies, La Jolla, CA, United States.
| |
Collapse
|
44
|
Vahid-Ansari F, Albert PR. Rewiring of the Serotonin System in Major Depression. Front Psychiatry 2021; 12:802581. [PMID: 34975594 PMCID: PMC8716791 DOI: 10.3389/fpsyt.2021.802581] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/17/2021] [Indexed: 12/14/2022] Open
Abstract
Serotonin is a key neurotransmitter that is implicated in a wide variety of behavioral and cognitive phenotypes. Originating in the raphe nuclei, 5-HT neurons project widely to innervate many brain regions implicated in the functions. During the development of the brain, as serotonin axons project and innervate brain regions, there is evidence that 5-HT plays key roles in wiring the developing brain, both by modulating 5-HT innervation and by influencing synaptic organization within corticolimbic structures. These actions are mediated by 14 different 5-HT receptors, with region- and cell-specific patterns of expression. More recently, the role of the 5-HT system in synaptic re-organization during adulthood has been suggested. The 5-HT neurons have the unusual capacity to regrow and reinnervate brain regions following insults such as brain injury, chronic stress, or altered development that result in disconnection of the 5-HT system and often cause depression, anxiety, and cognitive impairment. Chronic treatment with antidepressants that amplify 5-HT action, such as selective serotonin reuptake inhibitors (SSRIs), appears to accelerate the rewiring of the 5-HT system by mechanisms that may be critical to the behavioral and cognitive improvements induced in these models. In this review, we survey the possible 5-HT receptor mechanisms that could mediate 5-HT rewiring and assess the evidence that 5-HT-mediated brain rewiring is impacting recovery from mental illness. By amplifying 5-HT-induced rewiring processes using SSRIs and selective 5-HT agonists, more rapid and effective treatments for injury-induced mental illness or cognitive impairment may be achieved.
Collapse
Affiliation(s)
- Faranak Vahid-Ansari
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada
| |
Collapse
|
45
|
Biselli T, Lange SS, Sablottny L, Steffen J, Walther A. Optogenetic and chemogenetic insights into the neurocircuitry of depression-like behaviour: A systematic review. Eur J Neurosci 2021; 53:9-38. [PMID: 31633833 DOI: 10.1111/ejn.14603] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 09/13/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) and its treatment are challenges for global health. Optogenetics and chemogenetics are driving MDD research forward by unveiling causal relations between cell-type-specific control of neurons and depressive-like behaviour in rodents. Using a systematic search process, in this review, a set of 43 original studies applying optogenetic or chemogenetic techniques in rodent models of depression was identified. Our aim was to provide an examination of all available studies elucidating central neuronal mechanisms leading to depressive-like behaviour in rodents and thereby unveiling the most promising routes for future research. A complex interacting network of relevant structures, in which central circuitries causally related to depressive-like behaviour are implicated, has been identified. As most relevant structures emerge: medial prefrontal cortex, anterior cingulate cortex, amygdala, nucleus accumbens, ventral tegmental area, hippocampus and raphe nuclei. Further evidence, though examined by only few studies, emerges for structures like the lateral habenula, or medial dorsal thalamus. Most of the identified brain areas have previously been associated with MDD neuropathology, but now evidence can be provided for causal pathological mechanisms within a complex cortico-limbic reward circuitry. However, the studies also show conflicting results concerning the mechanisms underlying the causal involvement of specific circuitries. Comparability of studies is partly limited since even small deviations in methodological approaches lead to different outcomes. Factors influencing study outcomes were identified and need to be considered in future studies (e.g. frequency used for stimulation, time and duration of stimulation, limitations of applied animal models of MDD).
Collapse
Affiliation(s)
- Tom Biselli
- Biological Psychology, TU Dresden, Dresden, Germany
| | | | | | | | - Andreas Walther
- Biological Psychology, TU Dresden, Dresden, Germany
- Clinical Psychology and Psychotherapy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Papp M, Gruca P, Lason M, Litwa E, Solecki W, Willner P. AMPA receptors mediate the pro-cognitive effects of electrical and optogenetic stimulation of the medial prefrontal cortex in antidepressant non-responsive Wistar-Kyoto rats. J Psychopharmacol 2020; 34:1418-1430. [PMID: 33200659 PMCID: PMC7708672 DOI: 10.1177/0269881120967857] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND The chronic mild stress (CMS) procedure is a widely used animal model of depression, and its application in Wistar-Kyoto (WKY) rats has been validated as a model of antidepressant-refractory depression. While not responding to chronic treatment with antidepressant drugs, WKY rats do respond to acute deep brain stimulation (DBS) of the medial prefrontal cortex (mPFC). In antidepressant-responsive strains there is evidence suggesting a role for AMPA subtype of glutamate receptor in the action mechanism of both antidepressants and DBS. METHODS Animals were subjected to CMS for 6 to 8 weeks; sucrose intake was monitored weekly and novel object recognition (NOR) test was conducted following recovery from CMS. Wistars were treated chronically with venlafaxine (VEN), while WKY were treated acutely with either DBS, optogenetic stimulation (OGS) of virally-transduced (AAV5-hSyn-ChR2-EYFP) mPFC or ventral hippocampus, or acute intra-mPFC injection of the AMPA receptor positive allosteric modulator CX-516. The AMPA receptor antagonist NBQX was administered, at identical sites in mPFC, immediately following the exposure trial in the NOR. RESULTS Sucrose intake and NOR were suppressed by CMS, and restored by VEN in Wistars and by DBS, OGS, or CX-516 in WKY. However, OGS of the ventral hippocampal afferents to mPFC was ineffective. A low dose of NBQX selectively blocked the procognitive effect of VEN, DBS and OGS. CONCLUSIONS These results suggest that activation of AMPA receptors in the mPFC represents a common pathway for the antidepressant effects of both conventional (VEN) and novel (DBS, OGS) antidepressant modalities, in both antidepressant responsive (Wistar) and antidepressant-resistant (WKY) rats.
Collapse
Affiliation(s)
- Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland,Mariusz Papp, Maj Institute of Pharmacology Polish Academy of Sciences, 12 Smetna Street, Krakow, 31-343, Poland.
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Wojciech Solecki
- Department of Neurobiology and Neuropsychology, Institute of Applied Psychology, Jagiellonian University, Krakow, Poland
| | - Paul Willner
- Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
47
|
Calarco CA, Lobo MK. Depression and substance use disorders: Clinical comorbidity and shared neurobiology. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 157:245-309. [PMID: 33648671 DOI: 10.1016/bs.irn.2020.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mood disorders, including major depressive disorder (MDD), are the most prevalent psychiatric illnesses, and pose an incredible burden to society, both in terms of disability and in terms of costs associated with medical care and lost work time. MDD has extremely high rates of comorbidity with substance use disorders (SUD) as many of the same neurobiological circuits and molecular mechanisms regulate the reward pathways disrupted in both conditions. MDD may induce SUDs, SUD may contribute to MDD development, or underlying vulnerabilities and common life experience may confer risk to developing both conditions. In this chapter we explore theories of MDD and SUD comorbidity, the neurobiological underpinnings of depression, overlapping cellular and molecular pathways for both conditions, and current treatment approaches for these comorbid conditions.
Collapse
Affiliation(s)
- Cali A Calarco
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
48
|
Yuan Y, Li D, Yu F, Kang X, Xu H, Zhang P. Effects of Akt/mTOR/p70S6K Signaling Pathway Regulation on Neuron Remodeling Caused by Translocation Repair. Front Neurosci 2020; 14:565870. [PMID: 33132828 PMCID: PMC7550644 DOI: 10.3389/fnins.2020.565870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022] Open
Abstract
Peripheral nerve injury repair has been considered a difficult problem in the field of trauma for a long time. Conventional surgical methods are not applicable in some special types of nerve injury, prompting scholars to seek to develop more effective nerve translocation repair technologies. The purpose of this study was to explore the functional state of neurons in injured lower limbs after translocation repair, with a view to preliminarily clarify the molecular mechanisms underlying this process. Eighteen Sprague–Dawley rats were divided into the normal, tibial nerve in situ repair, and common peroneal nerve transposition repair tibial nerve groups. Nerve function assessment and immunohistochemical staining of neurofilament 200 (NF-200), protein kinase B (Akt), mammalian target of rapamycin (mTOR), and ribosomal protein S6 kinase (p70S6K) in the dorsal root ganglia were performed at 12 weeks after surgery. Tibial nerve function and neuroelectrophysiological analysis, osmic acid staining, muscle strength testing, and muscle fiber staining showed that the nerve translocation repair could restore the function of the recipient nerve to a certain extent; however, the repair was not as efficient as the in situ repair. Immunohistochemical staining showed that the translocation repair resulted in changes in the microstructure of neuronal cell bodies, and the expressions of Akt, mTOR, and p70S6K in the three dorsal root ganglia groups were significantly different (p < 0.05). This study demonstrates that the nerve translocation repair technology sets up a new reflex loop, with the corresponding neuroskeletal adjustments, in which, donor neurons dominate the recipient nerves. This indicates that nerve translocation repair technology can lead to neuronal remodeling and is important as a supplementary treatment for a peripheral nerve injury. Furthermore, the Akt/mTOR/p70S6K signaling pathway may be involved in the formation of the new neural reflex loop created as a result of the translocation repair.
Collapse
Affiliation(s)
- Yusong Yuan
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Dongdong Li
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Department of Orthopedics, PLA Strategic Support Force Characteristic Medical Center, Beijing, China
| | - Fei Yu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Xuejing Kang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| | - Hailin Xu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China.,Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Peixun Zhang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing, China.,Key Laboratory of Trauma and Neural Regeneration, Ministry of Education, Peking University, Beijing, China.,National Center for Trauma Medicine, Beijing, China
| |
Collapse
|
49
|
Xia F, Kheirbek MA. Circuit-Based Biomarkers for Mood and Anxiety Disorders. Trends Neurosci 2020; 43:902-915. [PMID: 32917408 PMCID: PMC7606349 DOI: 10.1016/j.tins.2020.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/23/2020] [Accepted: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Mood and anxiety disorders are complex heterogeneous syndromes that manifest in dysfunctions across multiple brain regions, cell types, and circuits. Biomarkers using brain-wide activity patterns in humans have proven useful in distinguishing between disorder subtypes and identifying effective treatments. In order to improve biomarker identification, it is crucial to understand the basic circuitry underpinning brain-wide activity patterns. Leveraging a large repertoire of techniques, animal studies have examined roles of specific cell types and circuits in driving maladaptive behavior. Recent advances in multiregion recording techniques, data-driven analysis approaches, and machine-learning-based behavioral analysis tools can further push the boundary of animal studies and bridge the gap with human studies, to assess how brain-wide activity patterns encode and drive emotional behavior. Together, these efforts will allow identifying more precise biomarkers to enhance diagnosis and treatment.
Collapse
Affiliation(s)
- Frances Xia
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| | - Mazen A Kheirbek
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA; Center for Integrative Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
50
|
Hare BD, Duman RS. Prefrontal cortex circuits in depression and anxiety: contribution of discrete neuronal populations and target regions. Mol Psychiatry 2020; 25:2742-2758. [PMID: 32086434 PMCID: PMC7442605 DOI: 10.1038/s41380-020-0685-9] [Citation(s) in RCA: 222] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/03/2020] [Accepted: 02/10/2020] [Indexed: 12/12/2022]
Abstract
Our understanding of depression and its treatment has advanced with the advent of ketamine as a rapid-acting antidepressant and the development and refinement of tools capable of selectively altering the activity of populations of neuronal subtypes. This work has resulted in a paradigm shift away from dysregulation of single neurotransmitter systems in depression towards circuit level abnormalities impacting function across multiple brain regions and neurotransmitter systems. Studies on the features of circuit level abnormalities demonstrate structural changes within the prefrontal cortex (PFC) and functional changes in its communication with distal brain structures. Treatments that impact the activity of brain regions, such as transcranial magnetic stimulation or rapid-acting antidepressants like ketamine, appear to reverse depression associated circuit abnormalities though the mechanisms underlying the reversal, as well as development of these abnormalities remains unclear. Recently developed optogenetic and chemogenetic tools that allow high-fidelity control of neuronal activity in preclinical models have begun to elucidate the contributions of the PFC and its circuitry to depression- and anxiety-like behavior. These tools offer unprecedented access to specific circuits and neuronal subpopulations that promise to offer a refined view of the circuit mechanisms surrounding depression and potential mechanistic targets for development and reversal of depression associated circuit abnormalities.
Collapse
Affiliation(s)
- Brendan D. Hare
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut,Corresponding author and lead contact:
| | - Ronald S. Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|