1
|
Chan M, Ogawa S. GPR139, an Ancient Receptor and an Emerging Target for Neuropsychiatric and Behavioral Disorders. Mol Neurobiol 2025:10.1007/s12035-025-04828-2. [PMID: 40102345 DOI: 10.1007/s12035-025-04828-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Accepted: 03/09/2025] [Indexed: 03/20/2025]
Abstract
GPR139 is an orphan G-protein-coupled receptor that is predominantly expressed in several midbrain regions, e.g., the habenula, striatum, and hypothalamus. GPR139 gene is highly conserved across vertebrate phylogenetic taxa, suggesting its fundamental importance in neurophysiology. Evidence from both animal studies and human genetic association studies has demonstrated that dysregulation of GPR139 expression and function is linked to aberrant behaviors, cognitive deficits, alterations in sleep and alertness, and substance abuse and withdrawal. Animal knockout models suggest that GPR139 plays an anti-opioid role by modulating the signaling activity of the μ-opioid receptor (MOR), as well as the intensity of withdrawal symptoms and nociception in behavioral paradigms. Modulation of GPR139 activity by surrogate agonists such as TAK-041 and JNJ-63533054 has shown promising results in experimental models; however, the use of TAK-041 in clinical trials has produced heterogeneous effects and has not met the intended primary endpoint. Here, we highlight current in vitro and in vivo studies of GPR139, its potential physiological roles, and therapeutic potential in the pathophysiology of neuropsychiatric and behavioral disorders. This review aims to focus on the current knowledge gaps to facilitate future studies that will contribute to the understanding of GPR139 as a therapeutic target for neuropsychiatric and behavioral disorders.
Collapse
Affiliation(s)
- Minyu Chan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia
| | - Satoshi Ogawa
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
2
|
Rahim AR, Will V, Myung J. Mood variation under dual regulation of circadian clock and light. Chronobiol Int 2025; 42:162-184. [PMID: 39840618 DOI: 10.1080/07420528.2025.2455144] [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: 09/16/2024] [Revised: 12/16/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
The intricate relationship between circadian rhythms and mood is well-established. Disturbances in circadian rhythms and sleep often precede the development of mood disorders, such as major depressive disorder (MDD), bipolar disorder (BD), and seasonal affective disorder (SAD). Two primary factors, intrinsic circadian clocks and light, drive the natural fluctuations in mood throughout the day, mirroring the patterns of sleepiness and wakefulness. Nearly all organisms possess intrinsic circadian clocks that coordinate daily rhythms, with light serving as the primary environmental cue to synchronize these internal timekeepers with the 24-hour cycle. Additionally, light directly influences mood states. Disruptions to circadian rhythms, such as those caused by jet lag, shift work, or reduced daylight hours, can trigger or exacerbate mood symptoms. The complex and often subtle connections between circadian disruptions and mood dysregulation suggest that focusing solely on individual clock genes is insufficient to fully understand their etiology and progression. Instead, mood instability may arise from systemic misalignments between external cycles and the internal synchronization of circadian clocks. Here, we synthesize past research on the independent contributions of circadian clocks and light to mood regulation, drawing particularly on insights from animal studies that illuminate fundamental mechanisms relevant to human health.
Collapse
Affiliation(s)
- Amalia Ridla Rahim
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
| | - Veronica Will
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
| | - Jihwan Myung
- Laboratory of Braintime, Graduate Institute of Mind, Brain and Consciousness (GIMBC), Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences (GIMS), Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
3
|
Loonen AJM. The putative role of the habenula in animal migration. Physiol Behav 2024; 286:114668. [PMID: 39151652 DOI: 10.1016/j.physbeh.2024.114668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/26/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND When an addicted animal seeks a specific substance, it is based on the perception of internal and external cues that strongly motivate to pursue the acquisition of that compound. In essence, a similar process acts out when an animal leaves its present area to begin its circannual migration. This review article examines the existence of scientific evidence for possible relatedness of migration and addiction by influencing Dorsal Diencephalic Conduction System (DDCS) including the habenula. METHODS For this review especially the databases of Pubmed and Embase were frequently and non-systematically searched. RESULTS The mechanisms of bird migration have been thoroughly investigated. Especially the mechanism of the circannual biorhythm and its associated endocrine regulation has been well elucidated. A typical behavior called "Zugunruhe" marks the moment of leaving in migratory birds. The role of magnetoreception in navigation has also been clarified in recent years. However, how bird migration is regulated at the neuronal level in the forebrain is not well understood. Among mammals, marine mammals are most similar to birds. They use terrestrial magnetic field when navigating and often bridge long distances between breeding and foraging areas. Population migration is further often seen among the large hoofed mammals in different parts of the world. Importantly, learning processes and social interactions with conspecifics play a major role in these ungulates. Considering the evolutionary development of the forebrain in vertebrates, it can be postulated that the DDCS plays a central role in regulating the readiness and intensity of essential (emotional) behaviors. There is manifold evidence that this DDCS plays an important role in relapse to abuse after prolonged periods of abstinence from addictive behavior. It is also possible that the DDCS plays a role in navigation. CONCLUSIONS The role of the DDCS in the neurobiological regulation of bird migration has hardly been investigated. The involvement of this system in relapse to addiction in mammals might suggest to change this. It is recommended that particularly during "Zugunruhe" the role of neuronal regulation via the DDCS will be further investigated.
Collapse
Affiliation(s)
- Anton J M Loonen
- Pharmacotherapy, Epidemiology & Economics, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, the Netherlands.
| |
Collapse
|
4
|
Narain P, Petković A, Šušić M, Haniffa S, Anwar M, Arnoux M, Drou N, Antonio-Saldi G, Chaudhury D. Nighttime-specific differential gene expression in suprachiasmatic nucleus and habenula is associated with resilience to chronic social stress. Transl Psychiatry 2024; 14:407. [PMID: 39358331 PMCID: PMC11447250 DOI: 10.1038/s41398-024-03100-w] [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: 02/16/2024] [Revised: 09/10/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
The molecular mechanisms that link stress and biological rhythms still remain unclear. The habenula (Hb) is a key brain region involved in regulating diverse types of emotion-related behaviours while the suprachiasmatic nucleus (SCN) is the body's central clock. To investigate the effects of chronic social stress on transcription patterns, we performed gene expression analysis in the Hb and SCN of stress-naïve and stress-exposed mice. Our analysis revealed a large number of differentially expressed genes and enrichment of synaptic and cell signalling pathways between resilient and stress-naïve mice at zeitgeber 16 (ZT16) in both the Hb and SCN. This transcriptomic signature was nighttime-specific and observed only in stress-resilient mice. In contrast, there were relatively few differences between the stress-susceptible and stress-naïve groups across time points. Our results reinforce the functional link between circadian gene expression patterns and differential responses to stress, thereby highlighting the importance of temporal expression patterns in homoeostatic stress responses.
Collapse
Affiliation(s)
- Priyam Narain
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Aleksa Petković
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marko Šušić
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Salma Haniffa
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Mariam Anwar
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Marc Arnoux
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Nizar Drou
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Dipesh Chaudhury
- Center for Genomics and Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Department of Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Center for Brain and Health, New York University Abu Dhabi, Abu Dhabi, UAE.
| |
Collapse
|
5
|
Chen X, Liu X, Luan S, Wang X, Zhang Y, Hao Y, Zhang Q, Zhang J, Zhao H. Optogenetic activation of the lateral habenula D1R-ventral tegmental area circuit induces depression-like behavior in mice. Eur Arch Psychiatry Clin Neurosci 2024; 274:867-878. [PMID: 38236282 DOI: 10.1007/s00406-023-01743-2] [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: 02/19/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
A number of different receptors are distributed in glutamatergic neurons of the lateral habenula (LHb). These glutamatergic neurons are involved in different neural pathways, which may identify how the LHb regulates various physiological functions. However, the role of dopamine D1 receptor (D1R)-expressing habenular neurons projecting to the ventral tegmental area (VTA) (LHbD1R-VTA) remains not well understood. In the current study, to determine the activity of D1R-expressing neurons in LHb, D1R-Cre mice were used to establish the chronic restraint stress (CRS) depression model. Adeno-associated virus was injected into bilateral LHb in D1R-Cre mice to examine whether optogenetic activation of the LHb D1R-expressing neurons and their projections could induce depression-like behavior. Optical fibers were implanted in the LHb and VTA, respectively. To investigate whether optogenetic inhibition of the LHbD1R-VTA circuit could produce antidepressant-like effects, the adeno-associated virus was injected into the bilateral LHb in the D1R-Cre CRS model, and optical fibers were implanted in the bilateral VTA. The D1R-expressing neuronal activity in the LHb was increased in the CRS depression model. Optogenetic activation of the D1R-expressing neurons in LHb induced behavioral despair and anhedonia, which could also be induced by activation of the LHbD1R-VTA axons. Conversely, optogenetic inhibition of the LHbD1R-VTA circuit improved behavioral despair and anhedonia in the CRS depression model. D1R-expressing glutamatergic neurons in the LHb and their projections to the VTA are involved in the occurrence and regulation of depressive-like behavior.
Collapse
Affiliation(s)
- Xiaowei Chen
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
- Department of Rehabilitation Medicine, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Xiaofeng Liu
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Shuxin Luan
- Department of Mental Health, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Xuxin Wang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
| | - Ying Zhang
- Department of Neurology, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yulei Hao
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Qiang Zhang
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China
| | - Jiaming Zhang
- Department of Rehabilitation Medicine, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Hua Zhao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Xinmin Street No. 126, Changchun, 130021, People's Republic of China.
- Neuroscience Research Center, First Hospital of Jilin University, Changchun, 130021, People's Republic of China.
| |
Collapse
|
6
|
van Rosmalen L, Deota S, Maier G, Le HD, Lin T, Ramasamy RK, Hut RA, Panda S. Energy balance drives diurnal and nocturnal brain transcriptome rhythms. Cell Rep 2024; 43:113951. [PMID: 38508192 PMCID: PMC11330649 DOI: 10.1016/j.celrep.2024.113951] [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: 10/04/2023] [Revised: 01/30/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Plasticity in daily timing of activity has been observed in many species, yet the underlying mechanisms driving nocturnality and diurnality are unknown. By regulating how much wheel-running activity will be rewarded with a food pellet, we can manipulate energy balance and switch mice to be nocturnal or diurnal. Here, we present the rhythmic transcriptome of 21 tissues, including 17 brain regions, sampled every 4 h over a 24-h period from nocturnal and diurnal male CBA/CaJ mice. Rhythmic gene expression across tissues comprised different sets of genes with minimal overlap between nocturnal and diurnal mice. We show that non-clock genes in the suprachiasmatic nucleus (SCN) change, and the habenula was most affected. Our results indicate that adaptive flexibility in daily timing of behavior is supported by gene expression dynamics in many tissues and brain regions, especially in the habenula, which suggests a crucial role for the observed nocturnal-diurnal switch.
Collapse
Affiliation(s)
- Laura van Rosmalen
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Shaunak Deota
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Geraldine Maier
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Terry Lin
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ramesh K Ramasamy
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Roelof A Hut
- Chronobiology Unit, Groningen Institute for Evolutionary Life Sciences, University of Groningen, 9700 CC Groningen, the Netherlands.
| | - Satchidananda Panda
- Regulatory Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
Pereira AR, Alemi M, Cerqueira-Nunes M, Monteiro C, Galhardo V, Cardoso-Cruz H. Dynamics of Lateral Habenula-Ventral Tegmental Area Microcircuit on Pain-Related Cognitive Dysfunctions. Neurol Int 2023; 15:1303-1319. [PMID: 37987455 PMCID: PMC10660716 DOI: 10.3390/neurolint15040082] [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: 09/19/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/22/2023] Open
Abstract
Chronic pain is a health problem that affects the ability to work and perform other activities, and it generally worsens over time. Understanding the complex pain interaction with brain circuits could help predict which patients are at risk of developing central dysfunctions. Increasing evidence from preclinical and clinical studies suggests that aberrant activity of the lateral habenula (LHb) is associated with depressive symptoms characterized by excessive negative focus, leading to high-level cognitive dysfunctions. The primary output region of the LHb is the ventral tegmental area (VTA), through a bidirectional connection. Recently, there has been growing interest in the complex interactions between the LHb and VTA, particularly regarding their crucial roles in behavior regulation and their potential involvement in the pathological impact of chronic pain on cognitive functions. In this review, we briefly discuss the structural and functional roles of the LHb-VTA microcircuit and their impact on cognition and mood disorders in order to support future studies addressing brain plasticity during chronic pain conditions.
Collapse
Affiliation(s)
- Ana Raquel Pereira
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Mobina Alemi
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Mariana Cerqueira-Nunes
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Programa Doutoral em Neurociências, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Clara Monteiro
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Vasco Galhardo
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Helder Cardoso-Cruz
- Instituto de Investigação e Inovação em Saúde—Pain Neurobiology Group, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (A.R.P.); (M.A.); (M.C.-N.); (C.M.); (V.G.)
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Departamento de Biomedicina—Unidade de Biologia Experimental, Faculdade de Medicina, Universidade do Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
8
|
Cincotta AH. Brain Dopamine-Clock Interactions Regulate Cardiometabolic Physiology: Mechanisms of the Observed Cardioprotective Effects of Circadian-Timed Bromocriptine-QR Therapy in Type 2 Diabetes Subjects. Int J Mol Sci 2023; 24:13255. [PMID: 37686060 PMCID: PMC10487918 DOI: 10.3390/ijms241713255] [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: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/27/2023] [Indexed: 09/10/2023] Open
Abstract
Despite enormous global efforts within clinical research and medical practice to reduce cardiovascular disease(s) (CVD), it still remains the leading cause of death worldwide. While genetic factors clearly contribute to CVD etiology, the preponderance of epidemiological data indicate that a major common denominator among diverse ethnic populations from around the world contributing to CVD is the composite of Western lifestyle cofactors, particularly Western diets (high saturated fat/simple sugar [particularly high fructose and sucrose and to a lesser extent glucose] diets), psychosocial stress, depression, and altered sleep/wake architecture. Such Western lifestyle cofactors are potent drivers for the increased risk of metabolic syndrome and its attendant downstream CVD. The central nervous system (CNS) evolved to respond to and anticipate changes in the external (and internal) environment to adapt survival mechanisms to perceived stresses (challenges to normal biological function), including the aforementioned Western lifestyle cofactors. Within the CNS of vertebrates in the wild, the biological clock circuitry surveils the environment and has evolved mechanisms for the induction of the obese, insulin-resistant state as a survival mechanism against an anticipated ensuing season of low/no food availability. The peripheral tissues utilize fat as an energy source under muscle insulin resistance, while increased hepatic insulin resistance more readily supplies glucose to the brain. This neural clock function also orchestrates the reversal of the obese, insulin-resistant condition when the low food availability season ends. The circadian neural network that produces these seasonal shifts in metabolism is also responsive to Western lifestyle stressors that drive the CNS clock into survival mode. A major component of this natural or Western lifestyle stressor-induced CNS clock neurophysiological shift potentiating the obese, insulin-resistant state is a diminution of the circadian peak of dopaminergic input activity to the pacemaker clock center, suprachiasmatic nucleus. Pharmacologically preventing this loss of circadian peak dopaminergic activity both prevents and reverses existing metabolic syndrome in a wide variety of animal models of the disorder, including high fat-fed animals. Clinically, across a variety of different study designs, circadian-timed bromocriptine-QR (quick release) (a unique formulation of micronized bromocriptine-a dopamine D2 receptor agonist) therapy of type 2 diabetes subjects improved hyperglycemia, hyperlipidemia, hypertension, immune sterile inflammation, and/or adverse cardiovascular event rate. The present review details the seminal circadian science investigations delineating important roles for CNS circadian peak dopaminergic activity in the regulation of peripheral fuel metabolism and cardiovascular biology and also summarizes the clinical study findings of bromocriptine-QR therapy on cardiometabolic outcomes in type 2 diabetes subjects.
Collapse
|
9
|
Ables JL, Park K, Ibañez-Tallon I. Understanding the habenula: A major node in circuits regulating emotion and motivation. Pharmacol Res 2023; 190:106734. [PMID: 36933754 PMCID: PMC11081310 DOI: 10.1016/j.phrs.2023.106734] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/04/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
Over the last decade, the understanding of the habenula has rapidly advanced from being an understudied brain area with the Latin name 'habena" meaning "little rein", to being considered a "major rein" in the control of key monoaminergic brain centers. This ancient brain structure is a strategic node in the information flow from fronto-limbic brain areas to brainstem nuclei. As such, it plays a crucial role in regulating emotional, motivational, and cognitive behaviors and has been implicated in several neuropsychiatric disorders, including depression and addiction. This review will summarize recent findings on the medial (MHb) and lateral (LHb) habenula, their topographical projections, cell types, and functions. Additionally, we will discuss contemporary efforts that have uncovered novel molecular pathways and synaptic mechanisms with a focus on MHb-Interpeduncular nucleus (IPN) synapses. Finally, we will explore the potential interplay between the habenula's cholinergic and non-cholinergic components in coordinating related emotional and motivational behaviors, raising the possibility that these two pathways work together to provide balanced roles in reward prediction and aversion, rather than functioning independently.
Collapse
Affiliation(s)
- Jessica L Ables
- Psychiatry Department, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kwanghoon Park
- The Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Inés Ibañez-Tallon
- The Laboratory of Molecular Biology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
10
|
Johnson TB, Brudvig JJ, Likhite S, Pratt MA, White KA, Cain JT, Booth CD, Timm DJ, Davis SS, Meyerink B, Pineda R, Dennys-Rivers C, Kaspar BK, Meyer K, Weimer JM. Early postnatal administration of an AAV9 gene therapy is safe and efficacious in CLN3 disease. Front Genet 2023; 14:1118649. [PMID: 37035740 PMCID: PMC10080320 DOI: 10.3389/fgene.2023.1118649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/02/2023] [Indexed: 04/11/2023] Open
Abstract
CLN3 disease, caused by biallelic mutations in the CLN3 gene, is a rare pediatric neurodegenerative disease that has no cure or disease modifying treatment. The development of effective treatments has been hindered by a lack of etiological knowledge, but gene replacement has emerged as a promising therapeutic platform for such disorders. Here, we utilize a mouse model of CLN3 disease to test the safety and efficacy of a cerebrospinal fluid-delivered AAV9 gene therapy with a study design optimized for translatability. In this model, postnatal day one administration of the gene therapy virus resulted in robust expression of human CLN3 throughout the CNS over the 24-month duration of the study. A range of histopathological and behavioral parameters were assayed, with the therapy consistently and persistently rescuing a number of hallmarks of disease while being safe and well-tolerated. Together, the results show great promise for translation of the therapy into the clinic, prompting the launch of a first-in-human clinical trial (NCT03770572).
Collapse
Affiliation(s)
- Tyler B. Johnson
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Jon J. Brudvig
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Shibi Likhite
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Melissa A. Pratt
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Katherine A. White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Jacob T. Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
| | - Clarissa D. Booth
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Derek J. Timm
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Samantha S. Davis
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Brandon Meyerink
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
| | - Ricardo Pineda
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | | | - Brian K. Kaspar
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Kathrin Meyer
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH, United States
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, United States
- Amicus Therapeutics, Cranbury, NJ, United States
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
11
|
Li Y, Ren J, Zhang Z, Weng Y, Zhang J, Zou X, Wu S, Hu H. Modification and Expression of mRNA m6A in the Lateral Habenular of Rats after Long-Term Exposure to Blue Light during the Sleep Period. Genes (Basel) 2023; 14:143. [PMID: 36672884 PMCID: PMC9859551 DOI: 10.3390/genes14010143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Artificial lighting, especially blue light, is becoming a public-health risk. Excessive exposure to blue light at night has been reported to be associated with brain diseases. However, the mechanisms underlying neuropathy induced by blue light remain unclear. An early anatomical tracing study described the projection of the retina to the lateral habenula (LHb), whereas more mechanistic reports are available on multiple brain functions and neuropsychiatric disorders in the LHb, which are rarely seen in epigenetic studies, particularly N6-methyladenosine (m6A). The purpose of our study was to first expose Sprague-Dawley rats to blue light (6.11 ± 0.05 mW/cm2, the same irradiance as 200 lx of white light in the control group) for 4 h, and simultaneously provide white light to the control group for the same time to enter a sleep period. The experiment was conducted over 12 weeks. RNA m6A modifications and different mRNA transcriptome profiles were observed in the LHb. We refer to this experimental group as BLS. High-throughput MeRIP-seq and mRNA-seq were performed, and we used bioinformatics to analyze the data. There were 188 genes in the LHb that overlapped between differentially m6A-modified mRNA and differentially expressed mRNA. The Kyoto Encyclopedia of Genes and Genomes and gene ontology analysis were used to enrich neuroactive ligand-receptor interaction, long-term depression, the cyclic guanosine monophosphate-dependent protein kinase G (cGMP-PKG) signaling pathway, and circadian entrainment. The m6A methylation level of the target genes in the BLS group was disordered. In conclusion, this study suggests that the mRNA expression and their m6A of the LHb were abnormal after blue light exposure during the sleep period, and the methylation levels of target genes related to synaptic plasticity were disturbed. This study offers a theoretical basis for the scientific use of light.
Collapse
Affiliation(s)
- Yinhan Li
- Fujian Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Jinjin Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350108, China
| | - Zhaoting Zhang
- School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Yali Weng
- School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Jian Zhang
- School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Xinhui Zou
- School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Siying Wu
- Fujian Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350108, China
| | - Hong Hu
- Fujian Key Laboratory of Environmental Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou 350108, China
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350108, China
| |
Collapse
|
12
|
Liu X, Huang H, Zhang Y, Wang L, Wang F. Sexual Dimorphism of Inputs to the Lateral Habenula in Mice. Neurosci Bull 2022; 38:1439-1456. [PMID: 35644002 PMCID: PMC9723051 DOI: 10.1007/s12264-022-00885-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/16/2022] [Indexed: 12/14/2022] Open
Abstract
The lateral habenula (LHb), which is a critical neuroanatomical hub and a regulator of midbrain monoaminergic centers, is activated by events resulting in negative valence and contributes to the expression of both appetitive and aversive behaviors. However, whole-brain cell-type-specific monosynaptic inputs to the LHb in both sexes remain incompletely elucidated. In this study, we used viral tracing combined with in situ hybridization targeting vesicular glutamate transporter 2 (vGlut2) and glutamic acid decarboxylase 2 (Gad2) to generate a comprehensive whole-brain atlas of inputs to glutamatergic and γ-aminobutyric acid (GABA)ergic neurons in the LHb. We found >30 ipsilateral and contralateral brain regions that projected to the LHb. Of these, there were significantly more monosynaptic LHb-projecting neurons from the lateral septum, anterior hypothalamus, dorsomedial hypothalamus, and ventromedial hypothalamus in females than in males. More interestingly, we found a stronger GABAergic projection from the medial septum to the LHb in males than in females. Our results reveal a comprehensive connectivity atlas of glutamatergic and GABAergic inputs to the LHb in both sexes, which may facilitate a better understanding of sexual dimorphism in physiological and pathological brain functions.
Collapse
Affiliation(s)
- Xue Liu
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongren Huang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yulin Zhang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Liping Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Feng Wang
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
| |
Collapse
|
13
|
Potential Role of the Circadian Clock in the Regulation of Cancer Stem Cells and Cancer Therapy. Int J Mol Sci 2022; 23:ijms232214181. [PMID: 36430659 PMCID: PMC9698777 DOI: 10.3390/ijms232214181] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
Circadian rhythms, including sleep/wake cycles as well as hormonal, immune, metabolic, and cell proliferation rhythms, are fundamental biological processes driven by a cellular time-keeping system called the circadian clock. Disruptions in these rhythms due to genetic alterations or irregular lifestyles cause fundamental changes in physiology, from metabolism to cellular proliferation and differentiation, resulting in pathological consequences including cancer. Cancer cells are not uniform and static but exist as different subtypes with phenotypic and functional differences in the tumor microenvironment. At the top of the heterogeneous tumor cell hierarchy, cancer stem cells (CSCs), a self-renewing and multi-potent cancer cell type, are most responsible for tumor recurrence and metastasis, chemoresistance, and mortality. Phenotypically, CSCs are associated with the epithelial-mesenchymal transition (EMT), which confers cancer cells with increased motility and invasion ability that is characteristic of malignant and drug-resistant stem cells. Recently, emerging studies of different cancer types, such as glioblastoma, leukemia, prostate cancer, and breast cancer, suggest that the circadian clock plays an important role in the maintenance of CSC/EMT characteristics. In this review, we describe recent discoveries regarding how tumor intrinsic and extrinsic circadian clock-regulating factors affect CSC evolution, highlighting the possibility of developing novel chronotherapeutic strategies that could be used against CSCs to fight cancer.
Collapse
|
14
|
Chrobok L, Ahern J, Piggins HD. Ticking and talking in the brainstem satiety centre: Circadian timekeeping and interactions in the diet-sensitive clock of the dorsal vagal complex. Front Physiol 2022; 13:931167. [PMID: 36117684 PMCID: PMC9481231 DOI: 10.3389/fphys.2022.931167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The dorsal vagal complex (DVC) is a key hub for integrating blood-borne, central, and vagal ascending signals that convey important information on metabolic and homeostatic state. Research implicates the DVC in the termination of food intake and the transition to satiety, and consequently it is considered a brainstem satiety centre. In natural and laboratory settings, animals have distinct times of the day or circadian phases at which they prefer to eat, but if and how circadian signals affect DVC activity is not well understood. Here, we evaluate how intrinsic circadian signals regulate molecular and cellular activity in the area postrema (AP), nucleus of the solitary tract (NTS), and dorsal motor nucleus of the vagus (DMV) of the DVC. The hierarchy and potential interactions among these oscillators and their response to changes in diet are considered a simple framework in which to model these oscillators and their interactions is suggested. We propose possible functions of the DVC in the circadian control of feeding behaviour and speculate on future research directions including the translational value of knowledge of intrinsic circadian timekeeping the brainstem.
Collapse
|
15
|
Suryadi, Cheng RK, Birkett E, Jesuthasan S, Chew LY. Dynamics and potential significance of spontaneous activity in the habenula. eNeuro 2022; 9:ENEURO.0287-21.2022. [PMID: 35981869 PMCID: PMC9450562 DOI: 10.1523/eneuro.0287-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
The habenula is an evolutionarily conserved structure of the vertebrate brain that is essential for behavioural flexibility and mood control. It is spontaneously active and is able to access diverse states when the animal is exposed to sensory stimuli. Here we investigate the dynamics of habenula spontaneous activity, to gain insight into how sensitivity is optimized. Two-photon calcium imaging was performed in resting zebrafish larvae at single cell resolution. An analysis of avalanches of inferred spikes suggests that the habenula is subcritical. Activity had low covariance and a small mean, arguing against dynamic criticality. A multiple regression estimator of autocorrelation time suggests that the habenula is neither fully asynchronous nor perfectly critical, but is reverberating. This pattern of dynamics may enable integration of information and high flexibility in the tuning of network properties, thus providing a potential mechanism for the optimal responses to a changing environment.Significance StatementSpontaneous activity in neurons shapes the response to stimuli. One structure with a high level of spontaneous neuronal activity is the habenula, a regulator of broadly acting neuromodulators involved in mood and learning. How does this activity influence habenula function? We show here that the habenula of a resting animal is near criticality, in a state termed reverberation. This pattern of dynamics is consistent with high sensitivity and flexibility, and may enable the habenula to respond optimally to a wide range of stimuli.
Collapse
Affiliation(s)
- Suryadi
- School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371
| | - Ruey-Kuang Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
| | - Elliot Birkett
- Institute of Molecular and Cell Biology, Singapore 138673
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom
| | - Suresh Jesuthasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921
- Institute of Molecular and Cell Biology, Singapore 138673
| | - Lock Yue Chew
- School of Physical & Mathematical Sciences, Nanyang Technological University, Singapore 637371
- Complexity Institute, Nanyang Technological University, Singapore 637335
| |
Collapse
|
16
|
Chen S, Sun X, Zhang Y, Mu Y, Su D. Habenula bibliometrics: Thematic development and research fronts of a resurgent field. Front Integr Neurosci 2022; 16:949162. [PMID: 35990593 PMCID: PMC9382245 DOI: 10.3389/fnint.2022.949162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/12/2022] [Indexed: 11/19/2022] Open
Abstract
The habenula (Hb) is a small structure of the posterior diencephalon that is highly conserved across vertebrates but nonetheless has attracted relatively little research attention until the past two decades. The resurgent interest is motivated by neurobehavioral studies demonstrating critical functions in a broad spectrum of motivational and cognitive processes, including functions relevant to psychiatric diseases. The Hb is widely conceived as an "anti-reward" center that acts by regulating brain monoaminergic systems. However, there is still no general conceptual framework for habenula research, and no study has focused on uncovering potentially significant but overlooked topics that may advance our understanding of physiological functions or suggest potential clinical applications of Hb-targeted interventions. Using science mapping tools, we quantitatively and qualitatively analyzed the relevant publications retrieved from the Web of Science Core Collection (WoSCC) database from 2002 to 2021. Herein we present an overview of habenula-related publications, reveal primary research trends, and prioritize some key research fronts by complementary bibliometric analysis. High-priority research fronts include Ventral Pallidum, Nucleus Accumbens, Nicotine and MHb, GLT-1, Zebrafish, and GCaMP, Ketamine, Deep Brain Stimulation, and GPR139. The high intrinsic heterogeneity of the Hb, extensive connectivity with both hindbrain and forebrain structures, and emerging associations with all three dimensions of mental disorders (internalizing, externalizing, and psychosis) suggest that the Hb may be the neuronal substrate for a common psychopathology factor shared by all mental illnesses termed the p factor. A future challenge is to explore the therapeutic potential of habenular modulation at circuit, cellular, and molecular levels.
Collapse
Affiliation(s)
- Sifan Chen
- Department of Anesthesiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyu Sun
- Department of Anesthesiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yizhe Zhang
- Department of Anesthesiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Mu
- State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Diansan Su
- Department of Anesthesiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
17
|
Pradel K, Drwięga G, Chrobok L, Błasiak T. Racing and Pacing in the Reward System: A Multi-Clock Circadian Control Over Dopaminergic Signalling. Front Physiol 2022; 13:932378. [PMID: 35812323 PMCID: PMC9259884 DOI: 10.3389/fphys.2022.932378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/07/2022] [Indexed: 11/27/2022] Open
Abstract
Level of motivation, responsiveness to rewards and punishment, invigoration of exploratory behaviours, and motor performance are subject to daily fluctuations that emerge from circadian rhythms in neuronal activity of the midbrain’s dopaminergic system. While endogenous circadian rhythms are weak in the ventral tegmental area and substantia nigra pars compacta, daily changes in expression of core clock genes, ion channels, neurotransmitter receptors, dopamine-synthesising enzymes, and dopamine transporters, accompanied by changes in electrical activity, are readily observed in these nuclei. These processes cause dopamine levels released in structures innervated by midbrain dopaminergic neurons (e.g., the striatum) to oscillate in a circadian fashion. Additionally, growing evidence show that the master circadian clock located in the suprachiasmatic nucleus of the hypothalamus (SCN) rhythmically influences the activity of the dopaminergic system through various intermediate targets. Thus, circadian changes in the activity of the dopaminergic system and concomitant dopamine release observed on a daily scale are likely to be generated both intrinsically and entrained by the master clock. Previous studies have shown that the information about the value and salience of stimuli perceived by the animal is encoded in the neuronal activity of brain structures innervating midbrain dopaminergic centres. Some of these structures themselves are relatively autonomous oscillators, while others exhibit a weak endogenous circadian rhythm synchronised by the SCN. Here, we place the dopaminergic system as a hub in the extensive network of extra-SCN circadian oscillators and discuss the possible consequences of its daily entrainment for animal physiology and behaviour.
Collapse
Affiliation(s)
- Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Gniewosz Drwięga
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
| | - Lukasz Chrobok
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, Bristol, United Kingdom
- *Correspondence: Lukasz Chrobok, ; Tomasz Błasiak,
| | - Tomasz Błasiak
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University, Kraków, Poland
- *Correspondence: Lukasz Chrobok, ; Tomasz Błasiak,
| |
Collapse
|
18
|
Yoo H, Kim HJ, Yang SH, Son GH, Gim JA, Lee HW, Kim H. Gene Expression Profiling of the Habenula in Rats Exposed to Chronic Restraint Stress. Mol Cells 2022; 45:306-316. [PMID: 35534192 PMCID: PMC9095505 DOI: 10.14348/molcells.2022.2257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 12/24/2022] Open
Abstract
Chronic stress contributes to the risk of developing depression; the habenula, a nucleus in epithalamus, is associated with many neuropsychiatric disorders. Using genome-wide gene expression analysis, we analyzed the transcriptome of the habenula in rats exposed to chronic restraint stress for 14 days. We identified 379 differentially expressed genes (DEGs) that were affected by chronic stress. These genes were enriched in neuroactive ligand-receptor interaction, the cAMP (cyclic adenosine monophosphate) signaling pathway, circadian entrainment, and synaptic signaling from the Kyoto Encyclopedia of Genes and Genomes pathway analysis and responded to corticosteroids, positive regulation of lipid transport, anterograde trans-synaptic signaling, and chemical synapse transmission from the Gene Ontology analysis. Based on protein-protein interaction network analysis of the DEGs, we identified neuroactive ligand-receptor interactions, circadian entrainment, and cholinergic synapse-related subclusters. Additionally, cell type and habenular regional expression of DEGs, evaluated using a recently published single-cell RNA sequencing study (GSE137478), strongly suggest that DEGs related to neuroactive ligand-receptor interaction and trans-synaptic signaling are highly enriched in medial habenular neurons. Taken together, our findings provide a valuable set of molecular targets that may play important roles in mediating the habenular response to stress and the onset of chronic stress-induced depressive behaviors.
Collapse
Affiliation(s)
- Hyeijung Yoo
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Biomedical Sciences, BrainKorea21 Four, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyun Jung Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Soo Hyun Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
| | - Gi Hoon Son
- Department of Legal Medicine, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyun Woo Lee
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Biomedical Sciences, BrainKorea21 Four, College of Medicine, Korea University, Seoul 02841, Korea
| | - Hyun Kim
- Department of Anatomy, College of Medicine, Korea University, Seoul 02841, Korea
- Department of Biomedical Sciences, BrainKorea21 Four, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
19
|
Becker-Krail DD, Walker WH, Nelson RJ. The Ventral Tegmental Area and Nucleus Accumbens as Circadian Oscillators: Implications for Drug Abuse and Substance Use Disorders. Front Physiol 2022; 13:886704. [PMID: 35574492 PMCID: PMC9094703 DOI: 10.3389/fphys.2022.886704] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/04/2022] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythms convergently evolved to allow for optimal synchronization of individuals’ physiological and behavioral processes with the Earth’s 24-h periodic cycling of environmental light and temperature. Whereas the suprachiasmatic nucleus (SCN) is considered the primary pacemaker of the mammalian circadian system, many extra-SCN oscillatory brain regions have been identified to not only exhibit sustainable rhythms in circadian molecular clock function, but also rhythms in overall region activity/function and mediated behaviors. In this review, we present the most recent evidence for the ventral tegmental area (VTA) and nucleus accumbens (NAc) to serve as extra-SCN oscillators and highlight studies that illustrate the functional significance of the VTA’s and NAc’s inherent circadian properties as they relate to reward-processing, drug abuse, and vulnerability to develop substance use disorders (SUDs).
Collapse
Affiliation(s)
- Darius D Becker-Krail
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - William H Walker
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Randy J Nelson
- Department of Neuroscience, Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
20
|
Depressive cognitive style relates to an individual trait of time perception in bipolar depression: a preliminary study. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2022. [DOI: 10.1016/j.jadr.2022.100363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
21
|
Hones VI, Mizumori SJY. Response Flexibility: The Role of the Lateral Habenula. Front Behav Neurosci 2022; 16:852235. [PMID: 35444521 PMCID: PMC9014270 DOI: 10.3389/fnbeh.2022.852235] [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: 01/11/2022] [Accepted: 03/01/2022] [Indexed: 01/13/2023] Open
Abstract
The ability to make appropriate decisions that result in an optimal outcome is critical for survival. This process involves assessing the environment as well as integrating prior knowledge about the environment with information about one's current internal state. There are many neural structures that play critical roles in mediating these processes, but it is not yet known how such information coalesces to influence behavioral output. The lateral habenula (LHb) has often been cited as a structure critical for adaptive and flexible responding when environmental contexts and internal state changes. A challenge, however, has been understanding how LHb promotes response flexibility. In this review, we hypothesize that the LHb enables flexible responding following the integration of context memory and internal state information by signaling downstream brainstem structures known to drive hippocampal theta. In this way, animals respond more flexibly in a task situation not because the LHb selects a particular action, but rather because LHb enhances a hippocampal neural state that is often associated with greater attention, arousal, and exploration. In freely navigating animals, these are essential conditions that are needed to discover and implement appropriate alternative choices and behaviors. As a corollary to our hypothesis, we describe short- and intermediate-term functions of the LHb. Finally, we discuss the effects on the behavior of LHb dysfunction in short- and intermediate-timescales, and then suggest that new therapies may act on the LHb to alleviate the behavioral impairments following long-term LHb disruption.
Collapse
Affiliation(s)
- Victoria I. Hones
- Department of Psychology, University of Washington, Seattle, WA, United States
| | - Sheri J. Y. Mizumori
- Department of Psychology, University of Washington, Seattle, WA, United States
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| |
Collapse
|
22
|
Baker PM, Mathis V, Lecourtier L, Simmons SC, Nugent FS, Hill S, Mizumori SJY. Lateral Habenula Beyond Avoidance: Roles in Stress, Memory, and Decision-Making With Implications for Psychiatric Disorders. Front Syst Neurosci 2022; 16:826475. [PMID: 35308564 PMCID: PMC8930415 DOI: 10.3389/fnsys.2022.826475] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 02/10/2022] [Indexed: 01/02/2023] Open
Abstract
In this Perspective review, we highlight some of the less explored aspects of lateral habenula (LHb) function in contextual memory, sleep, and behavioral flexibility. We provide evidence that LHb is well-situated to integrate different internal state and multimodal sensory information from memory-, stress-, motivational-, and reward-related circuits essential for both survival and decision making. We further discuss the impact of early life stress (ELS) on LHb function as an example of stress-induced hyperactivity and dysregulation of neuromodulatory systems within the LHb that promote anhedonia and motivational deficits following ELS. We acknowledge that recent technological advancements in manipulation and recording of neural circuits in simplified and well-controlled behavioral paradigms have been invaluable in our understanding of the critical role of LHb in motivation and emotional regulation as well as the involvement of LHb dysfunction in stress-induced psychopathology. However, we also argue that the use of ethologically-relevant behaviors with consideration of complex aspects of decision-making is warranted for future studies of LHb contributions in a wide range of psychiatric illnesses. We conclude this Perspective with some of the outstanding issues for the field to consider where a multi-systems approach is needed to investigate the complex nature of LHb circuitry interactions with environmental stimuli that predisposes psychiatric disorders.
Collapse
Affiliation(s)
- Phillip M. Baker
- Department of Psychology, Seattle Pacific University, Seattle, WA, United States
- *Correspondence: Phillip M. Baker,
| | - Victor Mathis
- CNRS UPR 3212, Institut des Neurosciences Cellulaires et Intégratives, Center National de la Recherche Scientifique, University of Strasbourg, Strasbourg, France
| | - Lucas Lecourtier
- CNRS, Laboratoire de Neurosciences Cognitives et Adaptatives (LNCA), UMR 7364, Université de Strasbourg, Strasbourg, France
- Lucas Lecourtier,
| | - Sarah C. Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Fereshteh S. Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Fereshteh S. Nugent,
| | - Sierra Hill
- Department of Psychology, Seattle Pacific University, Seattle, WA, United States
| | - Sheri J. Y. Mizumori
- Department of Psychology, University of Washington, Seattle, WA, United States
- Sheri J. Y. Mizumori,
| |
Collapse
|
23
|
Rapid-acting antidepressants and the circadian clock. Neuropsychopharmacology 2022; 47:805-816. [PMID: 34837078 PMCID: PMC8626287 DOI: 10.1038/s41386-021-01241-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/20/2021] [Accepted: 11/08/2021] [Indexed: 12/13/2022]
Abstract
A growing number of epidemiological and experimental studies has established that circadian disruption is strongly associated with psychiatric disorders, including major depressive disorder (MDD). This association is becoming increasingly relevant considering that modern lifestyles, social zeitgebers (time cues) and genetic variants contribute to disrupting circadian rhythms that may lead to psychiatric disorders. Circadian abnormalities associated with MDD include dysregulated rhythms of sleep, temperature, hormonal secretions, and mood which are modulated by the molecular clock. Rapid-acting antidepressants such as subanesthetic ketamine and sleep deprivation therapy can improve symptoms within 24 h in a subset of depressed patients, in striking contrast to conventional treatments, which generally require weeks for a full clinical response. Importantly, animal data show that sleep deprivation and ketamine have overlapping effects on clock gene expression. Furthermore, emerging data implicate the circadian system as a critical component involved in rapid antidepressant responses via several intracellular signaling pathways such as GSK3β, mTOR, MAPK, and NOTCH to initiate synaptic plasticity. Future research on the relationship between depression and the circadian clock may contribute to the development of novel therapeutic strategies for depression-like symptoms. In this review we summarize recent evidence describing: (1) how the circadian clock is implicated in depression, (2) how clock genes may contribute to fast-acting antidepressants, and (3) the mechanistic links between the clock genes driving circadian rhythms and neuroplasticity.
Collapse
|
24
|
Lefter R, Cojocariu RO, Ciobica A, Balmus IM, Mavroudis I, Kis A. Interactions between Sleep and Emotions in Humans and Animal Models. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:274. [PMID: 35208598 PMCID: PMC8877042 DOI: 10.3390/medicina58020274] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022]
Abstract
Recently, increased interest and efforts were observed in describing the possible interaction between sleep and emotions. Human and animal model studies addressed the implication of both sleep patterns and emotional processing in neurophysiology and neuropathology in suggesting a bidirectional interaction intimately modulated by complex mechanisms and factors. In this context, we aimed to discuss recent evidence and possible mechanisms implicated in this interaction, as provided by both human and animal models in studies. In addition, considering the affective component of brain physiological patterns, we aimed to find reasonable evidence in describing the two-way association between comorbid sleep impairments and psychiatric disorders. The main scientific literature databases (PubMed/Medline, Web of Science) were screened with keyword combinations for relevant content taking into consideration only English written papers and the inclusion and exclusion criteria, according to PRISMA guidelines. We found that a strong modulatory interaction between sleep processes and emotional states resides on the activity of several key brain structures, such as the amygdala, prefrontal cortex, hippocampus, and brainstem nuclei. In addition, evidence suggested that physiologically and behaviorally related mechanisms of sleep are intimately interacting with emotional perception and processing which could advise the key role of sleep in the unconscious character of emotional processes. However, further studies are needed to explain and correlate the functional analysis with causative and protective factors of sleep impairments and negative emotional modulation on neurophysiologic processing, mental health, and clinical contexts.
Collapse
Affiliation(s)
- Radu Lefter
- Center of Biomedical Research, Romanian Academy, Iasi Branch, B dul Carol I, no. 8, 700506 Iasi, Romania;
| | - Roxana Oana Cojocariu
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, no 11, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, B dul Carol I, no 8, 700505 Iasi, Romania
- Academy of Romanian Scientists, Splaiul Independentei nr. 54, Sector 5, 050094 Bucuresti, Romania
| | - Ioana-Miruna Balmus
- Department of Exact Sciences and Natural Sciences, Institute of Interdisciplinary Research, Alexandru Ioan Cuza University of Iasi, Alexandru Lapusneanu Street, no. 26, 700057 Iasi, Romania
| | - Ioannis Mavroudis
- Department of Neurology, Leeds Teaching Hospitals NHS Trust, Leeds LS2 9JT, UK;
| | - Anna Kis
- Institute of Cognitive Neuroscience and Psychology, Hungarian Academy of Sciences, 1117 Budapest, Hungary;
| |
Collapse
|
25
|
Young CJ, Lyons D, Piggins HD. Circadian Influences on the Habenula and Their Potential Contribution to Neuropsychiatric Disorders. Front Behav Neurosci 2022; 15:815700. [PMID: 35153695 PMCID: PMC8831701 DOI: 10.3389/fnbeh.2021.815700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
The neural circadian system consists of the master circadian clock in the hypothalamic suprachiasmatic nuclei (SCN) communicating time of day cues to the rest of the body including other brain areas that also rhythmically express circadian clock genes. Over the past 16 years, evidence has emerged to indicate that the habenula of the epithalamus is a candidate extra-SCN circadian oscillator. When isolated from the SCN, the habenula sustains rhythms in clock gene expression and neuronal activity, with the lateral habenula expressing more robust rhythms than the adjacent medial habenula. The lateral habenula is responsive to putative SCN output factors as well as light information conveyed to the perihabenula area. Neuronal activity in the lateral habenula is altered in depression and intriguingly disruptions in circadian rhythms can elevate risk of developing mental health disorders including depression. In this review, we will principally focus on how circadian and light signals affect the lateral habenula and evaluate the possibility that alteration in these influences contribute to mental health disorders.
Collapse
|
26
|
Gouveia FV, Ibrahim GM. Habenula as a Neural Substrate for Aggressive Behavior. Front Psychiatry 2022; 13:817302. [PMID: 35250669 PMCID: PMC8891498 DOI: 10.3389/fpsyt.2022.817302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/25/2022] [Indexed: 11/17/2022] Open
Abstract
Over the past decades, an ever growing body of literature has explored the anatomy, connections, and functions of the habenula (Hb). It has been postulated that the Hb plays a central role in the control of the monoaminergic system, thus influencing a wide range of behavioral responses, and participating in the pathophysiology of a number of psychiatric disorders and neuropsychiatric symptoms, such as aggressive behaviors. Aggressive behaviors are frequently accompanied by restlessness and agitation, and are commonly observed in patients with psychiatric disorders, intellectual disabilities, and neurodegenerative diseases of aging. Recently, the Hb has been explored as a new target for neuromodulation therapies, such as deep brain stimulation, with promising results. Here we review the anatomical organization of the habenula and discuss several distinct mechanisms by which the Hb is involved in the modulation of aggressive behaviors, and propose new investigations for the development of novel treatments targeting the habenula to reduce aggressive behaviors.
Collapse
Affiliation(s)
- Flavia Venetucci Gouveia
- Neuroscience and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - George M Ibrahim
- Neuroscience and Mental Health, Hospital for Sick Children Research Institute, Toronto, ON, Canada.,Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
27
|
Multi-Modal Regulation of Circadian Physiology by Interactive Features of Biological Clocks. BIOLOGY 2021; 11:biology11010021. [PMID: 35053019 PMCID: PMC8772734 DOI: 10.3390/biology11010021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/26/2022]
Abstract
The circadian clock is a fundamental biological timing mechanism that generates nearly 24 h rhythms of physiology and behaviors, including sleep/wake cycles, hormone secretion, and metabolism. Evolutionarily, the endogenous clock is thought to confer living organisms, including humans, with survival benefits by adapting internal rhythms to the day and night cycles of the local environment. Mirroring the evolutionary fitness bestowed by the circadian clock, daily mismatches between the internal body clock and environmental cycles, such as irregular work (e.g., night shift work) and life schedules (e.g., jet lag, mistimed eating), have been recognized to increase the risk of cardiac, metabolic, and neurological diseases. Moreover, increasing numbers of studies with cellular and animal models have detected the presence of functional circadian oscillators at multiple levels, ranging from individual neurons and fibroblasts to brain and peripheral organs. These oscillators are tightly coupled to timely modulate cellular and bodily responses to physiological and metabolic cues. In this review, we will discuss the roles of central and peripheral clocks in physiology and diseases, highlighting the dynamic regulatory interactions between circadian timing systems and multiple metabolic factors.
Collapse
|
28
|
Abstract
Circadian clocks are biological timing mechanisms that generate 24-h rhythms of physiology and behavior, exemplified by cycles of sleep/wake, hormone release, and metabolism. The adaptive value of clocks is evident when internal body clocks and daily environmental cycles are mismatched, such as in the case of shift work and jet lag or even mistimed eating, all of which are associated with physiological disruption and disease. Studies with animal and human models have also unraveled an important role of functional circadian clocks in modulating cellular and organismal responses to physiological cues (ex., food intake, exercise), pathological insults (e.g. virus and parasite infections), and medical interventions (e.g. medication). With growing knowledge of the molecular and cellular mechanisms underlying circadian physiology and pathophysiology, it is becoming possible to target circadian rhythms for disease prevention and treatment. In this review, we discuss recent advances in circadian research and the potential for therapeutic applications that take patient circadian rhythms into account in treating disease.
Collapse
Affiliation(s)
- Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington
| | - Jeffrey M. Field
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amita Sehgal
- Howard Hughes Medical Institute, Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
29
|
Roy N, Parhar I. Habenula orphan G-protein coupled receptors in the pathophysiology of fear and anxiety. Neurosci Biobehav Rev 2021; 132:870-883. [PMID: 34801259 DOI: 10.1016/j.neubiorev.2021.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 10/19/2022]
Abstract
The phasic emotion, fear, and the tonic emotion, anxiety, have been conventionally inspected in clinical frameworks to epitomize memory acquisition, storage, and retrieval. However, inappropriate expression of learned fear in a safe environment and its resistance to suppression is a cardinal feature of various fear-related disorders. A significant body of literature suggests the involvement of extra-amygdala circuitry in fear disorders. Consistent with this view, the present review underlies incentives for the association between the habenula and fear memory. G protein-coupled receptors (GPCRs) are important to understand the molecular mechanisms central to fear learning due to their neuromodulatory role. The efficacy of a pharmacological strategy aimed at exploiting habenular-GPCR desensitization machinery can serve as a therapeutic target combating the pathophysiology of fear disorders. Originating from this milieu, the conserved nature of orphan GPCRs in the brain, with some having the highest expression in the habenula can lead to recent endeavors in understanding its functionality in fear circuitry.
Collapse
Affiliation(s)
- Nisa Roy
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| | - Ishwar Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
30
|
Brain Clocks, Sleep, and Mood. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 34773227 DOI: 10.1007/978-3-030-81147-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
The suprachiasmatic nucleus houses the master clock, but the genes which encode the circadian clock components are also expressed throughout the brain. Here, we review how circadian clock transcription factors regulate neuromodulator systems such as histamine, dopamine, and orexin that promote arousal. These circadian transcription factors all lead to repression of the histamine, dopamine, and orexin systems during the sleep period, so ensuring integration with the ecology of the animal. If these transcription factors are deleted or mutated, in addition to the global disturbances in circadian rhythms, this causes a chronic up-regulation of neuromodulators leading to hyperactivity, elevated mood, and reduced sleep, which have been suggested to be states resembling mania.
Collapse
|
31
|
Chrobok L, Pradel K, Janik ME, Sanetra AM, Bubka M, Myung J, Ridla Rahim A, Klich JD, Jeczmien-Lazur JS, Palus-Chramiec K, Lewandowski MH. Intrinsic circadian timekeeping properties of the thalamic lateral geniculate nucleus. J Neurosci Res 2021; 99:3306-3324. [PMID: 34758124 DOI: 10.1002/jnr.24973] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 01/08/2023]
Abstract
Circadian rhythmicity in mammals is sustained by the central brain clock-the suprachiasmatic nucleus of the hypothalamus (SCN), entrained to the ambient light-dark conditions through a dense retinal input. However, recent discoveries of autonomous clock gene expression cast doubt on the supremacy of the SCN and suggest circadian timekeeping mechanisms devolve to local brain clocks. Here, we use a combination of molecular, electrophysiological, and optogenetic tools to evaluate intrinsic clock properties of the main retinorecipient thalamic center-the lateral geniculate nucleus (LGN) in male rats and mice. We identify the dorsolateral geniculate nucleus as a slave oscillator, which exhibits core clock gene expression exclusively in vivo. Additionally, we provide compelling evidence for intrinsic clock gene expression accompanied by circadian variation in neuronal activity in the intergeniculate leaflet and ventrolateral geniculate nucleus (VLG). Finally, our optogenetic experiments propose the VLG as a light-entrainable oscillator, whose phase may be advanced by retinal input at the beginning of the projected night. Altogether, this study for the first time demonstrates autonomous timekeeping mechanisms shaping circadian physiology of the LGN.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marcelina Elzbieta Janik
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Anna Magdalena Sanetra
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Amalia Ridla Rahim
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
32
|
The habenula clock influences response to a stressor. Neurobiol Stress 2021; 15:100403. [PMID: 34632007 PMCID: PMC8488752 DOI: 10.1016/j.ynstr.2021.100403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/12/2022] Open
Abstract
The response of an animal to a sensory stimulus depends on the nature of the stimulus and on expectations, which are mediated by spontaneous activity. Here, we ask how circadian variation in the expectation of danger, and thus the response to a potential threat, is controlled. We focus on the habenula, a mediator of threat response that functions by regulating neuromodulator release, and use zebrafish as the experimental system. Single cell transcriptomics indicates that multiple clock genes are expressed throughout the habenula, while quantitative in situ hybridization confirms that the clock oscillates. Two-photon calcium imaging indicates a circadian change in spontaneous activity of habenula neurons. To assess the role of this clock, a truncated clocka gene was specifically expressed in the habenula. This partially inhibited the clock, as shown by changes in per3 expression as well as altered day-night variation in dopamine, serotonin and acetylcholine levels. Behaviourally, anxiety-like responses evoked by an alarm pheromone were reduced. Circadian effects of the pheromone were disrupted, such that responses in the day resembled those at night. Behaviours that are regulated by the pineal clock and not triggered by stressors were unaffected. We suggest that the habenula clock regulates the expectation of danger, thus providing one mechanism for circadian change in the response to a stressor.
Collapse
|
33
|
Chrobok L, Jeczmien-Lazur JS, Bubka M, Pradel K, Klekocinska A, Klich JD, Ridla Rahim A, Myung J, Kepczynski M, Lewandowski MH. Daily coordination of orexinergic gating in the rat superior colliculus-Implications for intrinsic clock activities in the visual system. FASEB J 2021; 35:e21930. [PMID: 34533886 DOI: 10.1096/fj.202100779rr] [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: 05/12/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 01/07/2023]
Abstract
The orexinergic system delivers excitation for multiple brain centers to facilitate behavioral arousal, with its malfunction resulting in narcolepsy, somnolence, and notably, visual hallucinations. Since the circadian clock underlies the daily arousal, a timed coordination is expected between the orexin system and its target subcortical visual system, including the superior colliculus (SC). Here, we use a combination of electrophysiological, immunohistochemical, and molecular approaches across 24 h, together with the neuronal tract-tracing methods to investigate the daily coordination between the orexin system and the rodent SC. Higher orexinergic input was found to occur nocturnally in the superficial layers of the SC, in time for nocturnal silencing of spontaneous firing in this visual brain area. We identify autonomous daily and circadian expression of clock genes in the SC, which may underlie these day-night changes. Additionally, we establish the lateral hypothalamic origin of the orexin innervation to the SC and that the SC neurons robustly respond to orexin A via OX2 receptor in both excitatory and GABAA receptor-dependent inhibitory manners. Together, our evidence elucidates the combination of intrinsic and extrinsic clock mechanisms that shape the daily function of the visual layers of the SC.
Collapse
Affiliation(s)
- Lukasz Chrobok
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jagoda Stanislawa Jeczmien-Lazur
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Kamil Pradel
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Aleksandra Klekocinska
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Jasmin Daniela Klich
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| | - Amalia Ridla Rahim
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan.,Brain and Consciousness Research Centre, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| | - Mariusz Kepczynski
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University in Krakow, Krakow, Poland
| | - Marian Henryk Lewandowski
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
34
|
Quintela T, Furtado A, Duarte AC, Gonçalves I, Myung J, Santos CRA. The role of circadian rhythm in choroid plexus functions. Prog Neurobiol 2021; 205:102129. [PMID: 34343629 DOI: 10.1016/j.pneurobio.2021.102129] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/17/2022]
Abstract
For several years, a great effort has been devoted to understand how circadian oscillations in physiological processes are determined by the circadian clock system. This system is composed by the master clock at the suprachiasmatic nucleus which sets the pace and tunes peripheral clocks in several organs. It was recently demonstrated that the choroid plexus epithelial cells that compose the blood-cerebrospinal fluid barrier hold a circadian clock which might control their multiple functions with implications for the maintenance of brain homeostasis. However, the choroid plexus activities regulated by its inner clock are still largely unknown. In this review, we propose that several choroid plexus functions might be regulated by the circadian clock, alike in other tissues. We provide evidences that the timing of cerebrospinal fluid secretion, clearance of amyloid-beta peptides and xenobiotics, and the barrier function of the blood-cerebrospinal fluid barrier are regulated by the circadian clock. These data, highlight that the circadian regulation of the blood-cerebrospinal fluid barrier must be taken into consideration for enhancing drug delivery to central nervous system disorders.
Collapse
Affiliation(s)
- Telma Quintela
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - André Furtado
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Jihwan Myung
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, No. 172-1 Sec. 2 Keelung Road, Da'an District, Taipei 106, Taiwan; Brain and Consciousness Research Centre, Shuang Ho Hospital, Ministry of Health and Welfare, No. 291 Zhongzheng Road, Zhonghe District, New Taipei City 235, Taiwan
| | - Cecília R A Santos
- CICS-UBI - Health Sciences Research Center, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
35
|
Lee YA, Goto Y. The Habenula in the Link Between ADHD and Mood Disorder. Front Behav Neurosci 2021; 15:699691. [PMID: 34248519 PMCID: PMC8264146 DOI: 10.3389/fnbeh.2021.699691] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a childhood-onset, neurodevelopmental disorder, whereas major depressive disorder (MDD) is a mood disorder that typically emerges in adulthood. Accumulating evidence suggests that these seemingly unrelated psychiatric disorders, whose symptoms even appear antithetical [e.g., psychomotor retardation in depression vs. hyperactivity (psychomotor acceleration) in ADHD], are in fact associated with each other. Thus, individuals with ADHD exhibit high comorbidity with MDD later in life. Moreover, genetic studies have shown substantial overlaps of susceptibility genes between ADHD and MDD. Here, we propose a novel and testable hypothesis that the habenula, the epithalamic brain region important for the regulation of monoamine transmission, may be involved in both ADHD and MDD. The hypothesis suggests that an initially hypoactive habenula during childhood in individuals with ADHD may undergo compensatory changes during development, priming the habenula to be hyperactive in response to stress exposure and thereby increasing vulnerability to MDD in adulthood. Moreover, we propose a new perspective on habenular deficits in psychiatric disorders that consider the habenula a neural substrate that could explain multiple psychiatric disorders.
Collapse
Affiliation(s)
- Young-A Lee
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| | - Yukiori Goto
- Primate Research Institute, Kyoto University, Inuyama, Japan
| |
Collapse
|
36
|
Costa R. Frontiers in Chronobiology: Endogenous Clocks at the Core of Signaling Pathways in Physiology. Front Physiol 2021; 12:684745. [PMID: 34093241 PMCID: PMC8173170 DOI: 10.3389/fphys.2021.684745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/14/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Rodolfo Costa
- Department of Biology, University of Padova, Italian National Research Council (CNR) Institute of Neuroscience, Padova, Italy
| |
Collapse
|
37
|
Liu H, Rastogi A, Narain P, Xu Q, Sabanovic M, Alhammadi AD, Guo L, Cao JL, Zhang H, Aqel H, Mlambo V, Rezgui R, Radwan B, Chaudhury D. Blunted diurnal firing in lateral habenula projections to dorsal raphe nucleus and delayed photoentrainment in stress-susceptible mice. PLoS Biol 2021; 19:e3000709. [PMID: 33690628 PMCID: PMC7984642 DOI: 10.1371/journal.pbio.3000709] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/22/2021] [Accepted: 02/04/2021] [Indexed: 01/29/2023] Open
Abstract
Daily rhythms are disrupted in patients with mood disorders. The lateral habenula (LHb) and dorsal raphe nucleus (DRN) contribute to circadian timekeeping and regulate mood. Thus, pathophysiology in these nuclei may be responsible for aberrations in daily rhythms during mood disorders. Using the 15-day chronic social defeat stress (CSDS) paradigm and in vitro slice electrophysiology, we measured the effects of stress on diurnal rhythms in firing of LHb cells projecting to the DRN (cellsLHb→DRN) and unlabeled DRN cells. We also performed optogenetic experiments to investigate if increased firing in cellsLHb→DRN during exposure to a weak 7-day social defeat stress (SDS) paradigm induces stress-susceptibility. Last, we investigated whether exposure to CSDS affected the ability of mice to photoentrain to a new light–dark (LD) cycle. The cellsLHb→DRN and unlabeled DRN cells of stress-susceptible mice express greater blunted diurnal firing compared to stress-näive (control) and stress-resilient mice. Daytime optogenetic activation of cellsLHb→DRN during SDS induces stress-susceptibility which shows the direct correlation between increased activity in this circuit and putative mood disorders. Finally, we found that stress-susceptible mice are slower, while stress-resilient mice are faster, at photoentraining to a new LD cycle. Our findings suggest that exposure to strong stressors induces blunted daily rhythms in firing in cellsLHb→DRN, DRN cells and decreases the initial rate of photoentrainment in susceptible-mice. In contrast, resilient-mice may undergo homeostatic adaptations that maintain daily rhythms in firing in cellsLHb→DRN and also show rapid photoentrainment to a new LD cycle. Daily rhythms are disrupted in patients suffering from mood disorders, and it is known that the lateral habenula and dorsal raphe nucleus contribute to circadian timekeeping and regulate mood. This study shows that stress-susceptible mice have blunted and inverted diurnal firing rhythms in lateral habenula cells that project to the dorsal raphe nucleus, and have a slow rate of photoentrainment to a new light cycle.
Collapse
Affiliation(s)
- He Liu
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Province Key Laboratory of Anesthesiology & Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, The Xuzhou Medical University, Xuzhou, China
| | - Ashutosh Rastogi
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Priyam Narain
- Center for Genomics and Systems Biology, New York University Abu Dhabi, United Arab Emirates
| | - Qing Xu
- Center for Genomics and Systems Biology, New York University Abu Dhabi, United Arab Emirates
| | - Merima Sabanovic
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | | | - Lihua Guo
- Center for Genomics and Systems Biology, New York University Abu Dhabi, United Arab Emirates
| | - Jun-Li Cao
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Center for Genomics and Systems Biology, New York University Abu Dhabi, United Arab Emirates
| | - Hongxing Zhang
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hala Aqel
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Vongai Mlambo
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Rachid Rezgui
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Basma Radwan
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Dipesh Chaudhury
- The Division of Science, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
- * E-mail:
| |
Collapse
|
38
|
Falup-Pecurariu C, Diaconu Ș, Țînț D, Falup-Pecurariu O. Neurobiology of sleep (Review). Exp Ther Med 2021; 21:272. [PMID: 33603879 DOI: 10.3892/etm.2021.9703] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Sleep is a physiological global state composed of two different phases: Non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. The control mechanisms of sleep manifest at the level of genetic, biological and cellular organization. Several brain areas, including the basal forebrain, thalamus, and hypothalamus, take part in regulating the activity of this status of life. The signals between different brain regions and those from cortical areas to periphery are conducted through various neuromediators, which are known to either promote wakefulness or sleep. Among others, serotonin, norepinephrine, histamine, hypocretin (orexin), acetylcholine, dopamine, glutamate, and gamma-aminobutyric acid are known to orchestrate the intrinsic mechanisms of sleep neurobiology. Several models that explain the transition and the continuity between wakefulness, NREM sleep and REM sleep have been proposed. All of these models include neurotransmitters as ligands in a complex reciprocal connectivity across the key-centers taking part in the regulation of sleep. Moreover, various environmental cues are integrated by a central pacemaker-located in the suprachiasmatic nucleus-which is able to connect with cortical regions and with peripheral tissues in order to promote the sleep-wake pattern.
Collapse
Affiliation(s)
- Cristian Falup-Pecurariu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Department of Neurology, County Emergency Clinic Hospital, 500365 Brașov, Romania
| | - Ștefania Diaconu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Department of Neurology, County Emergency Clinic Hospital, 500365 Brașov, Romania
| | - Diana Țînț
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania.,Clinicco Hospital, 500059 Brașov, Romania
| | - Oana Falup-Pecurariu
- Department of Neurology, Faculty of Medicine, Transilvania University of Brașov, 500036 Brașov, Romania
| |
Collapse
|
39
|
Ramaswamy M, Cheng RK, Jesuthasan S. Identification of GABAergic neurons innervating the zebrafish lateral habenula. Eur J Neurosci 2020; 52:3918-3928. [PMID: 32464693 PMCID: PMC7689879 DOI: 10.1111/ejn.14843] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/01/2022]
Abstract
Habenula neurons are constantly active. The level of activity affects mood and behaviour, with increased activity in the lateral habenula reflecting exposure to punishment and a switch to passive coping and depression. Here, we identify GABAergic neurons that could reduce activity in the lateral habenula of larval zebrafish. GAD65/67 immunohistochemistry and imaging of gad1b:DsRed transgenic fish suggest the presence of GABAergic terminals in the neuropil and between cell bodies in the lateral habenula. Retrograde tracing with the lipophilic dye DiD suggests that the former derives from the thalamus, while the latter originates from a group of cells in the posterior hypothalamus that are located between the posterior tuberal nucleus and hypothalamic lobes. Two‐photon calcium imaging indicates that blue light causes excitation of thalamic GABAergic neurons and terminals in the neuropil, while a subpopulation of lateral habenula neurons show reduced intracellular calcium levels. Whole‐cell electrophysiological recording indicates that blue light reduces membrane potential of lateral habenula neurons. These observations suggest that GABAergic input from the thalamus may mediate inhibition in the zebrafish lateral habenula. Mechanisms governing release of GABA from the neurons in the posterior hypothalamus, which are likely to be in the tuberomammillary nucleus, remain to be defined.
Collapse
Affiliation(s)
- Mahathi Ramaswamy
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Ruey-Kuang Cheng
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Suresh Jesuthasan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore.,Institute of Molecular and Cell Biology, Singapore
| |
Collapse
|
40
|
Begemann K, Neumann A, Oster H. Regulation and function of extra-SCN circadian oscillators in the brain. Acta Physiol (Oxf) 2020; 229:e13446. [PMID: 31965726 DOI: 10.1111/apha.13446] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022]
Abstract
Most organisms evolved endogenous, so called circadian clocks as internal timekeeping mechanisms allowing them to adapt to recurring changes in environmental demands brought about by 24-hour rhythms such as the light-dark cycle, temperature variations or changes in humidity. The mammalian circadian clock system is based on cellular oscillators found in all tissues of the body that are organized in a hierarchical fashion. A master pacemaker located in the suprachiasmatic nucleus (SCN) synchronizes peripheral tissue clocks and extra-SCN oscillators in the brain with each other and with external time. Different time cues (so called Zeitgebers) such as light, food intake, activity and hormonal signals reset the clock system through the SCN or by direct action at the tissue clock level. While most studies on non-SCN clocks so far have focused on peripheral tissues, several extra-SCN central oscillators were characterized in terms of circadian rhythm regulation and output. Some of them are directly innervated by the SCN pacemaker, while others receive indirect input from the SCN via other neural circuits or extra-brain structures. The specific physiological function of these non-SCN brain oscillators as well as their role in the regulation of the circadian clock network remains understudied. In this review we summarize our current knowledge about the regulation and function of extra-SCN circadian oscillators in different brain regions and devise experimental approaches enabling us to unravel the organization of the circadian clock network in the central nervous system.
Collapse
Affiliation(s)
| | | | - Henrik Oster
- Institute of Neurobiology University of Lübeck Lübeck Germany
| |
Collapse
|
41
|
Hu H, Cui Y, Yang Y. Circuits and functions of the lateral habenula in health and in disease. Nat Rev Neurosci 2020; 21:277-295. [PMID: 32269316 DOI: 10.1038/s41583-020-0292-4] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2020] [Indexed: 12/14/2022]
Abstract
The past decade has witnessed exponentially growing interest in the lateral habenula (LHb) owing to new discoveries relating to its critical role in regulating negatively motivated behaviour and its implication in major depression. The LHb, sometimes referred to as the brain's 'antireward centre', receives inputs from diverse limbic forebrain and basal ganglia structures, and targets essentially all midbrain neuromodulatory systems, including the noradrenergic, serotonergic and dopaminergic systems. Its unique anatomical position enables the LHb to act as a hub that integrates value-based, sensory and experience-dependent information to regulate various motivational, cognitive and motor processes. Dysfunction of the LHb may contribute to the pathophysiology of several psychiatric disorders, especially major depression. Recently, exciting progress has been made in identifying the molecular and cellular mechanisms in the LHb that underlie negative emotional state in animal models of drug withdrawal and major depression. A future challenge is to translate these advances into effective clinical treatments.
Collapse
Affiliation(s)
- Hailan Hu
- Department of Psychiatry of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. .,The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, Mental Health Center, Zhejiang University, Hangzhou, China. .,Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, China. .,Fountain-Valley Institute for Life Sciences, Guangzhou, China.
| | - Yihui Cui
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yan Yang
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| |
Collapse
|
42
|
Liu H, Rastogi A, Sabanovic M, Alhammadi AD, Xu Q, Guo L, Cao J, Zhang H, Narain P, Aqel H, Mlambo V, Rezgui R, Radwan B, Chaudhury D. Blunted Diurnal Firing in Lateral Habenula Projections to Dorsal Raphe Nucleus and Delayed Photoentrainment in Stress-Susceptible Mice.. [DOI: 10.1101/2020.03.19.998732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
Abstract
ABSTRACTDaily rhythms are disrupted in patients suffering from mood disorders. The lateral habenula (LHb) and dorsal raphe nucleus (DRN) contribute to circadian timekeeping and regulate mood. Thus, pathophysiology in these nuclei may be responsible for aberrations in daily rhythms during mood disorders. Using the chronic social defeat stress (CSDS) paradigm and in-vitro slice electrophysiology we measured the effects of stress on diurnal rhythms in firing of LHb cells projecting to the DRN (cellsLHb→DRN) and DRN cells alone. We also performed optogenetic experiments to investigate if increased firing in cellsLHb→DRN during exposure to subthreshold social defeat stress (SSDS), induces stress-susceptibility. Last we investigated whether exposure to CSDS affected the ability of mice to phototentrain to a new LD cycle. The cellsLHb→DRN and DRN cells alone of stress-susceptible mice express greater blunted diurnal firing compared to stress-naive (control) and stress-resilient mice. Day-time optogenetic activation of cellsLHb→DRN during SSDS induces stress-susceptibility which shows the direct correlation between increased activity in this circuit and putative mood disorders. Finally, we found that stress-susceptible mice are slower, while stress-resilient mice are faster, at photoentraining to a new LD cycle. Our findings suggest that CSDS induces blunted daily rhythms in firing in cellsLHb→DRN and slow rate of photoentrainment in susceptible-mice. In contrast, resilientmice may undergo homeostatic adaptations that maintain daily rhythms in firing in cellsLHb→DRN and also show rapid photoentrainment to a new LD-cycle.
Collapse
|
43
|
Zhao YN, Yan YD, Wang CY, Qu WM, Jhou TC, Huang ZL, Yang SR. The Rostromedial Tegmental Nucleus: Anatomical Studies and Roles in Sleep and Substance Addictions in Rats and Mice. Nat Sci Sleep 2020; 12:1215-1223. [PMID: 33380853 PMCID: PMC7769149 DOI: 10.2147/nss.s278026] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
The rostromedial tegmental nucleus (RMTg), a brake of the dopamine system, is specifically activated by aversive stimuli, such as foot shock. It is principally composed of gamma-aminobutyric acid neurons. However, there is no exact location of the RMTg on the brain stereotaxic atlas. The RMTg can be defined by c-Fos staining elicited by psychostimulants, the position of retrograde-labeled neurons stained by injections into the ventral tegmental area (VTA), the terminal field formed by axons from the lateral habenula, and some molecular markers identified as specifically expressed in the RMTg such as FoxP1. The RMTg receives a broad range of inputs and produces diverse outputs, which indicates that the RMTg has multiple functions. First, the RMTg plays an essential role for non-rapid eye movement sleep. Additionally, the RMTg serves a vital role in response to addiction. Opiates increase the firing rates of dopaminergic neurons in the VTA by acting on μ-opioid receptors on RMTg neurons and their terminals inside the VTA. In this review, we summarize the recent research advances on the anatomical location of the RMTg in rats and mice, its projections, and its regulation of sleep-wake behavior and addiction.
Collapse
Affiliation(s)
- Ya-Nan Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Yu-Dong Yan
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Chen-Yao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Wei-Min Qu
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Thomas C Jhou
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Zhi-Li Huang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| | - Su-Rong Yang
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
44
|
Lee HJ. Is Advancing Circadian Rhythm the Mechanism of Antidepressants? Psychiatry Investig 2019; 16:479-483. [PMID: 31352729 PMCID: PMC6664215 DOI: 10.30773/pi.2019.06.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 06/20/2019] [Indexed: 12/15/2022] Open
Abstract
Antidepressants usually require 2-8 weeks after drug administration to obtain a clinical response. In contrast, three fast-acting antidepressant treatments (sleep deprivation, electroconvulsive therapy, and ketamine) significantly reduced depressive symptoms within hours to days in a subgroup of patients with depressive disorder. This review addresses the mechanisms underlying these fast effects, with specific focus on treatment effects on circadian rhythms. Numerous recent studies have shown that circadian dysregulation may play an important role in the pathogenesis of mood disorders. These studies indicate that a common therapeutic mechanism underlying the three fast antidepressant therapies is related to circadian rhythm. Evidence suggests that depressive disorder is associated with circadian rhythm delay and that the mechanism of the antidepressant effect is a process in which the delayed circadian rhythm is restored to normal by the treatment.
Collapse
Affiliation(s)
- Heon-Jeong Lee
- Department of Psychiatry and Chronobiology Institute, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Paul JR, Davis JA, Goode LK, Becker BK, Fusilier A, Meador-Woodruff A, Gamble KL. Circadian regulation of membrane physiology in neural oscillators throughout the brain. Eur J Neurosci 2019; 51:109-138. [PMID: 30633846 DOI: 10.1111/ejn.14343] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/21/2022]
Abstract
Twenty-four-hour rhythmicity in physiology and behavior are driven by changes in neurophysiological activity that vary across the light-dark and rest-activity cycle. Although this neural code is most prominent in neurons of the primary circadian pacemaker in the suprachiasmatic nucleus (SCN) of the hypothalamus, there are many other regions in the brain where region-specific function and behavioral rhythmicity may be encoded by changes in electrical properties of those neurons. In this review, we explore the existing evidence for molecular clocks and/or neurophysiological rhythms (i.e., 24 hr) in brain regions outside the SCN. In addition, we highlight the brain regions that are ripe for future investigation into the critical role of circadian rhythmicity for local oscillators. For example, the cerebellum expresses rhythmicity in over 2,000 gene transcripts, and yet we know very little about how circadian regulation drives 24-hr changes in the neural coding responsible for motor coordination. Finally, we conclude with a discussion of how our understanding of circadian regulation of electrical properties may yield insight into disease mechanisms which may lead to novel chronotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Jodi R Paul
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jennifer A Davis
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lacy K Goode
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bryan K Becker
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Allison Fusilier
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Aidan Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Karen L Gamble
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
46
|
Collins B, Brown SA. Beyond the molecular clock. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
47
|
Belle MDC, Diekman CO. Neuronal oscillations on an ultra-slow timescale: daily rhythms in electrical activity and gene expression in the mammalian master circadian clockwork. Eur J Neurosci 2018; 48:2696-2717. [PMID: 29396876 DOI: 10.1111/ejn.13856] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/16/2018] [Accepted: 01/28/2018] [Indexed: 12/17/2022]
Abstract
Neuronal oscillations of the brain, such as those observed in the cortices and hippocampi of behaving animals and humans, span across wide frequency bands, from slow delta waves (0.1 Hz) to ultra-fast ripples (600 Hz). Here, we focus on ultra-slow neuronal oscillators in the hypothalamic suprachiasmatic nuclei (SCN), the master daily clock that operates on interlocking transcription-translation feedback loops to produce circadian rhythms in clock gene expression with a period of near 24 h (< 0.001 Hz). This intracellular molecular clock interacts with the cell's membrane through poorly understood mechanisms to drive the daily pattern in the electrical excitability of SCN neurons, exhibiting an up-state during the day and a down-state at night. In turn, the membrane activity feeds back to regulate the oscillatory activity of clock gene programs. In this review, we emphasise the circadian processes that drive daily electrical oscillations in SCN neurons, and highlight how mathematical modelling contributes to our increasing understanding of circadian rhythm generation, synchronisation and communication within this hypothalamic region and across other brain circuits.
Collapse
Affiliation(s)
- Mino D C Belle
- Institute of Clinical and Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, EX4 4PS, UK
| | - Casey O Diekman
- Department of Mathematical Sciences, New Jersey Institute of Technology, Newark, NJ, USA.,Institute for Brain and Neuroscience Research, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|
48
|
Circadian Rhythm Disturbances in Mood Disorders: Insights into the Role of the Suprachiasmatic Nucleus. Neural Plast 2017; 2017:1504507. [PMID: 29230328 PMCID: PMC5694588 DOI: 10.1155/2017/1504507] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/05/2017] [Accepted: 10/03/2017] [Indexed: 12/28/2022] Open
Abstract
Circadian rhythm disturbances are a common symptom among individuals with mood disorders. The suprachiasmatic nucleus (SCN), in the ventral part of the anterior hypothalamus, orchestrates physiological and behavioral circadian rhythms. The SCN consists of self-sustaining oscillators and receives photic and nonphotic cues, which entrain the SCN to the external environment. In turn, through synaptic and hormonal mechanisms, the SCN can drive and synchronize circadian rhythms in extra-SCN brain regions and peripheral tissues. Thus, genetic or environmental perturbations of SCN rhythms could disrupt brain regions more closely related to mood regulation and cause mood disturbances. Here, we review clinical and preclinical studies that provide evidence both for and against a causal role for the SCN in mood disorders.
Collapse
|