1
|
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
|
2
|
Cai J, Jiang Y, Xu Y, Jiang Z, Young C, Li H, Ortiz-Guzman J, Zhuo Y, Li Y, Xu Y, Arenkiel BR, Tong Q. An excitatory projection from the basal forebrain to the ventral tegmental area that underlies anorexia-like phenotypes. Neuron 2024; 112:458-472.e6. [PMID: 38056455 PMCID: PMC10922337 DOI: 10.1016/j.neuron.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/04/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Maladaptation in balancing internal energy needs and external threat cues may result in eating disorders. However, brain mechanisms underlying such maladaptations remain elusive. Here, we identified that the basal forebrain (BF) sends glutamatergic projections to glutamatergic neurons in the ventral tegmental area (VTA) in mice. Glutamatergic neurons in both regions displayed correlated responses to various stressors. Notably, in vivo manipulation of BF terminals in the VTA revealed that the glutamatergic BF → VTA circuit reduces appetite, increases locomotion, and elicits avoidance. Consistently, activation of VTA glutamatergic neurons reduced body weight, blunted food motivation, and caused hyperactivity with behavioral signs of anxiety, all hallmarks of typical anorexia symptoms. Importantly, activation of BF glutamatergic terminals in the VTA reduced dopamine release in the nucleus accumbens. Collectively, our results point to overactivation of the glutamatergic BF → VTA circuit as a potential cause of anorexia-like phenotypes involving reduced dopamine release.
Collapse
Affiliation(s)
- Jing Cai
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Yanyan Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yuanzhong Xu
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiying Jiang
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Claire Young
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hongli Li
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Yizhou Zhuo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Benjamin R Arenkiel
- Department of Molecular and Human Genetics and Department of Neuroscience, Baylor College of Medicine, and Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| | - Qingchun Tong
- Brown Foundation of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center & UTHealth Graduate School for Biomedical Sciences, University of Texas Health Science at Houston, Houston, TX 77030, USA; Department of Neurobiology and Anatomy of McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Ahn DB, Jang HB, Ryu Y, Kim HK, Guan X, Fan Y, Lee BH, Kim HY. A hypothalamus-habenula circuit regulates psychomotor responses induced by cocaine. Addict Biol 2023; 28:e13354. [PMID: 38017642 DOI: 10.1111/adb.13354] [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: 04/25/2023] [Revised: 09/04/2023] [Accepted: 10/16/2023] [Indexed: 11/30/2023]
Abstract
Administration of cocaine increases synaptic dopamine levels by blocking dopamine reuptake and leads to increased locomotor activity and compulsive drug-seeking behaviour. It has been suggested that the lateral hypothalamus (LH) or lateral habenula (LHb) is involved in drug-seeking behaviours. To explore the role of the LH and the LHb in cocaine-induced psychomotor responses, we tested whether modulation of the LH or the LH-LHb circuit affects cocaine-induced locomotion. Cocaine-induced locomotor activity and dopamine release were suppressed by the activation of the LH with 2-[2,6-difluoro-4-[[2-[(phenylsulfonyl)amino]ethyl]thio]phenoxy]acetamide (PEPA), an AMPA receptor agonist. When the LH was inhibited by microinjection of a GABA receptor agonists mixture prior to cocaine injection, the cocaine's effects were enhanced. Furthermore, optogenetic activation of the LH-LHb circuit attenuated the cocaine-induced locomotion, while optogenetic inhibition of the LH-LHb circuit increased it. In vivo extracellular recording found that the LH sent a glutamatergic projection to the LHb. These findings suggest that the LH glutamatergic projection to the LHb plays an active role in the modulation of cocaine-induced psychomotor responses.
Collapse
Affiliation(s)
- Dan Bi Ahn
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Han Byeol Jang
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, South Korea
| | - Yeonhee Ryu
- Korean Medicine Fundamental Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Hyung Kyu Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bae Hwan Lee
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
4
|
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
|
5
|
Du Y, Zhou S, Ma C, Chen H, Du A, Deng G, Liu Y, Tose AJ, Sun L, Liu Y, Wu H, Lou H, Yu YQ, Zhao T, Lammel S, Duan S, Yang H. Dopamine release and negative valence gated by inhibitory neurons in the laterodorsal tegmental nucleus. Neuron 2023; 111:3102-3118.e7. [PMID: 37499661 DOI: 10.1016/j.neuron.2023.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/25/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023]
Abstract
GABAergic neurons in the laterodorsal tegmental nucleus (LDTGABA) encode aversion by directly inhibiting mesolimbic dopamine (DA). Yet, the detailed cellular and circuit mechanisms by which these cells relay unpleasant stimuli to DA neurons and regulate behavioral output remain largely unclear. Here, we show that LDTGABA neurons bidirectionally respond to rewarding and aversive stimuli in mice. Activation of LDTGABA neurons promotes aversion and reduces DA release in the lateral nucleus accumbens. Furthermore, we identified two molecularly distinct LDTGABA cell populations. Somatostatin-expressing (Sst+) LDTGABA neurons indirectly regulate the mesolimbic DA system by disinhibiting excitatory hypothalamic neurons. In contrast, Reelin-expressing LDTGABA neurons directly inhibit downstream DA neurons. The identification of separate GABAergic subpopulations in a single brainstem nucleus that relay unpleasant stimuli to the mesolimbic DA system through direct and indirect projections is critical for establishing a circuit-level understanding of how negative valence is encoded in the mammalian brain.
Collapse
Affiliation(s)
- Yonglan Du
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Siyao Zhou
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Chenyan Ma
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Hui Chen
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Ana Du
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Guochuang Deng
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yige Liu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; College of Forensic Science, School of Medicine, Xi'an Jiaotong University, No.76, Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Amanda J Tose
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Li Sun
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yijun Liu
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Hangjun Wu
- Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou 310058, China
| | - Huifang Lou
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yan-Qin Yu
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Ting Zhao
- PKU-Nanjing Joint Institute of Translational Medicine, Nanjing 211800, China
| | - Stephan Lammel
- Division of Neurobiology, Department of Molecular and Cell Biology and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Shumin Duan
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Hongbin Yang
- Department of Affiliated Mental Health Center of Hangzhou Seventh People's Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, 1369 West Wenyi Road, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
6
|
Jang HB, Ahn D, Chang S, Kim HK, Lee BH, Kim SC, Steffensen SC, Bills KB, Lee H, Kim HY. Activation of a hypothalamus-habenula circuit by mechanical stimulation inhibits cocaine addiction-like behaviors. Biol Res 2023; 56:25. [PMID: 37194106 DOI: 10.1186/s40659-023-00440-7] [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: 09/27/2022] [Accepted: 05/11/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Mechanoreceptor activation modulates GABA neuron firing and dopamine (DA) release in the mesolimbic DA system, an area implicated in reward and substance abuse. The lateral habenula (LHb), the lateral hypothalamus (LH), and the mesolimbic DA system are not only reciprocally connected, but also involved in drug reward. We explored the effects of mechanical stimulation (MS) on cocaine addiction-like behaviors and the role of the LH-LHb circuit in the MS effects. MS was performed over ulnar nerve and the effects were evaluated by using drug seeking behaviors, optogenetics, chemogenetics, electrophysiology and immunohistochemistry. RESULTS Mechanical stimulation attenuated locomotor activity in a nerve-dependent manner and 50-kHz ultrasonic vocalizations (USVs) and DA release in nucleus accumbens (NAc) following cocaine injection. The MS effects were ablated by electrolytic lesion or optogenetic inhibition of LHb. Optogenetic activation of LHb suppressed cocaine-enhanced 50 kHz USVs and locomotion. MS reversed cocaine suppression of neuronal activity of LHb. MS also inhibited cocaine-primed reinstatement of drug-seeking behavior, which was blocked by chemogenetic inhibition of an LH-LHb circuit. CONCLUSION These findings suggest that peripheral mechanical stimulation activates LH-LHb pathways to attenuate cocaine-induced psychomotor responses and seeking behaviors.
Collapse
Affiliation(s)
- Han Byeol Jang
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - DanBi Ahn
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - Suchan Chang
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - Hyung Kyu Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Bong Hyo Lee
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu, 42158, South Korea
| | - Sang Chan Kim
- Medical Research Center, College of Korean Medicine, Daegu Haany University, Gyeongsan, 38610, South Korea
| | - Scott C Steffensen
- Department of Psychology and Neuroscience, Brigham Young University, Provo, UT, 84602, USA
| | - Kyle B Bills
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, UT, 84606, USA
| | - Hubert Lee
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
7
|
Noritake A, Nakamura K. Rewarding-unrewarding prediction signals under a bivalent context in the primate lateral hypothalamus. Sci Rep 2023; 13:5926. [PMID: 37045876 PMCID: PMC10097697 DOI: 10.1038/s41598-023-33026-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/06/2023] [Indexed: 04/14/2023] Open
Abstract
Animals can expect rewards under equivocal situations. The lateral hypothalamus (LH) is thought to process motivational information by producing valence signals of reward and punishment. Despite rich studies using rodents and non-human primates, these signals have been assessed separately in appetitive and aversive contexts; therefore, it remains unclear what information the LH encodes in equivocal situations. To address this issue, macaque monkeys were conditioned under a bivalent context in which reward and punishment were probabilistically delivered, in addition to appetitive and aversive contexts. The monkeys increased approaching behavior similarly in the bivalent and appetitive contexts as the reward probability increased. They increased avoiding behavior under the bivalent and aversive contexts as the punishment probability increased, but the mean frequency was lower under the bivalent context than under the aversive context. The population activity correlated with these mean behaviors. Moreover, the LH produced fine prediction signals of reward expectation, uncertainty, and predictability consistently in the bivalent and appetitive contexts by recruiting context-independent and context-dependent subpopulations of neurons, while it less produced punishment signals in the aversive and bivalent contexts. Further, neural ensembles encoded context information and "rewarding-unrewarding" and "reward-punishment" valence. These signals may motivate individuals robustly in equivocal environments.
Collapse
Affiliation(s)
- Atsushi Noritake
- Division of Behavioral Development, Department of System Neuroscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, 444-8585, Japan.
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Hayama, 240-0193, Japan.
| | - Kae Nakamura
- Department of Physiology, Kansai Medical University, 2-5-1, Shinmachi, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|
8
|
Thompson SM. Plasticity of synapses and reward circuit function in the genesis and treatment of depression. Neuropsychopharmacology 2023; 48:90-103. [PMID: 36057649 PMCID: PMC9700729 DOI: 10.1038/s41386-022-01422-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/08/2022]
Abstract
What changes in brain function cause the debilitating symptoms of depression? Can we use the answers to this question to invent more effective, faster acting antidepressant drug therapies? This review provides an overview and update of the converging human and preclinical evidence supporting the hypothesis that changes in the function of excitatory synapses impair the function of the circuits they are embedded in to give rise to the pathological changes in mood, hedonic state, and thought processes that characterize depression. The review also highlights complementary human and preclinical findings that classical and novel antidepressant drugs relieve the symptoms of depression by restoring the functions of these same synapses and circuits. These findings offer a useful path forward for designing better antidepressant compounds.
Collapse
Affiliation(s)
- Scott M Thompson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, 80045, CO, USA.
| |
Collapse
|
9
|
Lee SM, Jang HB, Fan Y, Lee BH, Kim SC, Bills KB, Steffensen SC, Kim HY. Nociceptive Stimuli Activate the Hypothalamus-Habenula Circuit to Inhibit the Mesolimbic Reward System and Cocaine-Seeking Behaviors. J Neurosci 2022; 42:9180-9192. [PMID: 36280259 PMCID: PMC9761669 DOI: 10.1523/jneurosci.0577-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 01/07/2023] Open
Abstract
Nociceptive signals interact with various regions of the brain, including those involved in physical sensation, reward, cognition, and emotion. Emerging evidence points to a role of nociception in the modulation of the mesolimbic reward system. The mechanism by which nociception affects dopamine (DA) signaling and reward is unclear. The lateral hypothalamus (LH) and the lateral habenula (LHb) receive somatosensory inputs and are structurally connected with the mesolimbic DA system. Here, we show that the LH-LHb pathway is necessary for nociceptive modulation of this system using male Sprague Dawley rats. Our extracellular single-unit recordings and head-mounted microendoscopic calcium imaging revealed that nociceptive stimulation by tail pinch excited LHb and LH neurons, which was inhibited by chemical lesion of the LH. Tail pinch increased activity of GABA neurons in ventral tegmental area, decreased the extracellular DA level in the nucleus accumbens ventrolateral shell in intact rats, and reduced cocaine-increased DA concentration, which was blocked by disruption of the LH. Furthermore, tail pinch attenuated cocaine-induced locomotor activity, 22 and 50 kHz ultrasonic vocalizations, and reinstatement of cocaine-seeking behavior, which was inhibited by chemogenetic silencing of the LH-LHb pathway. Our findings suggest that nociceptive stimulation recruits the LH-LHb pathway to inhibit mesolimbic DA system and drug reinstatement.SIGNIFICANCE STATEMENT The LHb and the LH have been implicated in processing nociceptive signals and modulating DA release in the mesolimbic DA system. Here, we show that the LH-LHb pathway is critical for nociception-induced modulation of mesolimbic DA release and cocaine reinstatement. Nociceptive stimulation alleviates extracellular DA release in the mesolimbic DA system, cocaine-induced psychomotor activities, and reinstatement of cocaine-seeking behaviors through the LH-LHb pathway. These findings provide novel evidence for sensory modulation of the mesolimbic DA system and drug addiction.
Collapse
Affiliation(s)
- Soo Min Lee
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu 41062, South Korea
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu 42158, South Korea
| | - Han Byeol Jang
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu 42158, South Korea
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu 42158, South Korea
| | - Bong Hyo Lee
- Department of Physiology, College of Korean Medicine, Daegu Haany University, Daegu 42158, South Korea
| | - Sang Chan Kim
- Medical Research Center, College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, South Korea
| | - Kyle B Bills
- Department of Biomedical Sciences, Noorda College of Osteopathic Medicine, Provo, Utah 84606
| | - Scott C Steffensen
- Department of Psychology and Neuroscience, Brigham Young University, Provo, Utah 84602
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, South Korea
| |
Collapse
|
10
|
Barcomb K, Olah SS, Kennedy MJ, Ford CP. Properties and modulation of excitatory inputs to the locus coeruleus. J Physiol 2022; 600:4897-4916. [PMID: 36156249 PMCID: PMC9669264 DOI: 10.1113/jp283605] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/22/2022] [Indexed: 01/12/2023] Open
Abstract
Excitatory inputs drive burst firing of locus coeruleus (LC) noradrenaline (NA) neurons in response to a variety of stimuli. Though a small number of glutamatergic LC afferents have been investigated, the overall landscape of these excitatory inputs is largely unknown. The current study used an optogenetic approach to isolate three glutamatergic afferents: the prefrontal cortex (PFC), lateral hypothalamus (LH) and periaqueductal grey (PAG). AAV5-DIO-ChR2 was injected into each region in male and female CaMKII-Cre mice and the properties of excitatory inputs on LC-NA cells were measured. Notably we found differences among these inputs. First, the pattern of axonal innervation differed between inputs such that LH afferents were concentrated in the posterior portion of the LC-NA somatic region while PFC afferents were denser in the medial dendritic region. Second, basal intrinsic properties varied for afferents, with LH inputs having the highest connectivity and the largest amplitude excitatory postsynaptic currents while PAG inputs had the lowest initial release probability. Third, while orexin and oxytocin had minimal effects on any input, dynorphin strongly inhibited excitatory inputs originating from the LH and PAG, and corticotrophin releasing factor (CRF) selectively inhibited inputs from the PAG. Overall, these results demonstrate that individual afferents to the LC have differing properties, which may contribute to the modularity of the LC and its ability to mediate various behavioural outcomes. KEY POINTS: Excitatory inputs to the locus coeruleus (LC) are important for driving noradrenaline neuron activity and downstream behaviours in response to salient stimuli, but little is known about the functional properties of different glutamate inputs that innervate these neurons We used a virus-mediated optogenetic approach to compare glutamate afferents from the prefrontal cortex (PFC), the lateral hypothalamus (LH) and the periaqueductal grey (PAG). While PFC was predicted to make synaptic inputs, we found that the LH and PAG also drove robust excitatory events in LC noradrenaline neurons. The strength, kinetics, and short-term plasticity of each input differed as did the extent of neuromodulation by both dynorphin and corticotrophin releasing factor. Thus each input displayed a unique set of basal properties and modulation by peptides. This characterization is an important step in deciphering the heterogeneity of the LC.
Collapse
Affiliation(s)
- Kelsey Barcomb
- Department of PharmacologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Samantha S. Olah
- Department of PharmacologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Matthew J. Kennedy
- Department of PharmacologyUniversity of Colorado School of MedicineAuroraCOUSA
| | - Christopher P. Ford
- Department of PharmacologyUniversity of Colorado School of MedicineAuroraCOUSA
| |
Collapse
|
11
|
Stria medullaris innervation follows the transcriptomic division of the habenula. Sci Rep 2022; 12:10118. [PMID: 35710872 PMCID: PMC9203815 DOI: 10.1038/s41598-022-14328-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
The habenula is a complex neuronal population integrated in a pivotal functional position into the vertebrate limbic system. Its main afference is the stria medullaris and its main efference the fasciculus retroflexus. This neuronal complex is composed by two main components, the medial and lateral habenula. Transcriptomic and single cell RNAseq studies have unveiled the morphological complexity of both components. The aim of our work was to analyze the relation between the origin of the axonal fibers and their final distribution in the habenula. We analyzed 754 tracing experiments from Mouse Brain Connectivity Atlas, Allen Brain Map databases, and selected 12 neuronal populations projecting into the habenular territory. Our analysis demonstrated that the projections into the medial habenula discriminate between the different subnuclei and are generally originated in the septal territory. The innervation of the lateral habenula displayed instead a less restricted distribution from preoptic, terminal hypothalamic and peduncular nuclei. Only the lateral oval subnucleus of the lateral habenula presented a specific innervation from the dorsal entopeduncular nucleus. Our results unveiled the necessity of novel sorts of behavioral experiments to dissect the different functions associated with the habenular complex and their correlation with the distinct neuronal populations that generate them.
Collapse
|
12
|
Xiang X, Chen Y, Li KX, Fang J, Bickler PE, Guan Z, Zhou W. Neuroanatomical Basis for the Orexinergic Modulation of Anesthesia Arousal and Pain Control. Front Cell Neurosci 2022; 16:891631. [PMID: 35558876 PMCID: PMC9090436 DOI: 10.3389/fncel.2022.891631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Hypothalamic orexin (hypocretin) neurons play crucial roles in arousal control. Their involvement in anesthesia and analgesia remains to be better understood. In order to enhance our view on the neuroanatomy, we systematically mapped the projections of orexin neurons with confocal microscope and light sheet microscope. We specifically expressed optogenetic opsins tagged with fluorescence markers in orexin neurons through adeno-associated viral infection in the mouse brain. The imaging results revealed fine details and novel features of the orexin projections throughout the brain, particularly related to the nuclei regulating arousal and pain. We then optogenetically activated orexin neurons in the lateral hypothalamus to study the effects on anesthesia-related behaviors. cFos staining showed that optogenetic stimulation can activate orexin neurons in the ChR2-mCherry group, but not the control mCherry group (62.86 ± 3.923% vs. 7.9 ± 2.072%; P < 0.0001). In behavior assays, optogenetic stimulation in the ChR2-mCherry group consistently elicited robust arousal from light isoflurane anesthesia (9.429 ± 3.804 s vs. 238.2 ± 17.42 s; P < 0.0001), shortened the emergence time after deep isoflurane anesthesia (109.5 ± 13.59 s vs. 213.8 ± 21.77 s; P = 0.0023), and increased the paw withdrawal latency in a hotplate test (11.45 ± 1.185 s vs. 8.767 ± 0.7775; P = 0.0317). The structural details of orexin fibers established the neuroanatomic basis for studying the role of orexin in anesthesia and analgesia.
Collapse
Affiliation(s)
- Xuaner Xiang
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Yuzhang Chen
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Ke-Xin Li
- Department of Physiology, University of California, San Francisco, San Francisco, CA, United States
| | - Jianqiao Fang
- Key Laboratory of Acupuncture and Neurology of Zhejiang Province, The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Philip E. Bickler
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
| | - Wei Zhou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, United States
- *Correspondence: Wei Zhou,
| |
Collapse
|
13
|
Barbier M, Croizier S, Alvarez-Bolado G, Risold PY. The distribution of Dlx1-2 and glutamic acid decarboxylase in the embryonic and adult hypothalamus reveals three differentiated LHA subdivisions in rodents. J Chem Neuroanat 2022; 121:102089. [DOI: 10.1016/j.jchemneu.2022.102089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 01/15/2022] [Accepted: 03/08/2022] [Indexed: 11/28/2022]
|
14
|
A diencephalic circuit in rats for opioid analgesia but not positive reinforcement. Nat Commun 2022; 13:764. [PMID: 35140231 PMCID: PMC8828762 DOI: 10.1038/s41467-022-28332-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/17/2022] [Indexed: 12/21/2022] Open
Abstract
Mu opioid receptor (MOR) agonists are potent analgesics, but also cause sedation, respiratory depression, and addiction risk. The epithalamic lateral habenula (LHb) signals aversive states including pain, and here we found that it is a potent site for MOR-agonist analgesia-like responses in rats. Importantly, LHb MOR activation is not reinforcing in the absence of noxious input. The LHb receives excitatory inputs from multiple sites including the ventral tegmental area, lateral hypothalamus, entopeduncular nucleus, and the lateral preoptic area of the hypothalamus (LPO). Here we report that LHb-projecting glutamatergic LPO neurons are excited by noxious stimulation and are preferentially inhibited by MOR selective agonists. Critically, optogenetic stimulation of LHb-projecting LPO neurons produces an aversive state that is relieved by LHb MOR activation, and optogenetic inhibition of LHb-projecting LPO neurons relieves the aversiveness of ongoing pain. Opioids are potent analgesics but also have addiction risk. Here a lateral preoptic area to lateral habenula connection is identified by which opioids relieve ongoing pain but do not produce reward in animals that do not have ongoing pain.
Collapse
|
15
|
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: 1.0] [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
|
16
|
Gakare SG, Varghese SS, Patni PP, Wagh SA, Ugale RR. Prevention of glutamate excitotoxicity in lateral habenula alleviates ethanol withdrawal-induced somatic and behavioral effects in ethanol dependent mice. Behav Brain Res 2022; 416:113557. [PMID: 34453973 DOI: 10.1016/j.bbr.2021.113557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 08/23/2021] [Accepted: 08/23/2021] [Indexed: 12/22/2022]
Abstract
Ethanol withdrawal commonly leads to anxiety-related disorder, a central factor toward negative reinforcement leading to relapse. The lateral habenula (LHb), an epithalamic nucleus, has emerged to be critical for both reward and aversion processing. Recent studies have also implicated the hyperactivity of LHb, adding to the emergence of negative emotional states during withdrawal from addictive drugs. Herein, we have studied the effects of glutamate transporter inhibitor (PDC), GluN2B-containing NMDAR antagonist (Ro25-6981), and intracellular calcium chelator (BAPTA-AM) injection in LHb on ethanol withdrawal symptoms. We found that ethanol 4 g/kg 20 % w/v intragastric (i.g.) for 10 days followed by 24 h of withdrawal showed a significant increase in somatic signs characterized by vocalization, shaking, and scratching. It also increased locomotor activity and anxiety-like behavior, collectively showing expression of ethanol withdrawal symptoms. The intra-LHb administration of PDC (0.5 ng) worsened the effect of ethanol withdrawal, whereas Ro25-6981 (2 and 4 ng) and BAPTA-AM (6.5 and 13 ng) significantly reversed ethanol withdrawal-induced behavior evident by a decrease in somatic signs, locomotor activity, and anxiety-like behavior. Further, pretreatment of Ro25-6981 and BAPTA-AM reduced the neuronal loss, whereas PDC increased it compared to the vehicle-treated group, as evidenced by NeuN staining. Altogether, our results suggest that increased glutamate, GluN2B activation, and likely calcium increase indicative of glutamate excitotoxicity-induced neuronal loss in LHb possibly endorse the emergence of ethanol withdrawal symptoms, while their inhibition might help in alleviating the ethanol withdrawal symptoms.
Collapse
Affiliation(s)
- Sukanya G Gakare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Shejin S Varghese
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Paras P Patni
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Samruddhi A Wagh
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India
| | - Rajesh R Ugale
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440 033, India.
| |
Collapse
|
17
|
Carlson HN, Christensen BA, Pratt WE. Stimulation of mu opioid, but not GABAergic, receptors of the lateral habenula alters free feeding in rats. Neurosci Lett 2021; 771:136417. [PMID: 34954115 DOI: 10.1016/j.neulet.2021.136417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 01/24/2023]
Abstract
Overconsumption, or eating beyond the point of homeostasis, is a key feature in the development of obesity. Although people are consuming beyond the point of homeostasis, they are not consuming constantly or indefinitely. Thus, there is likely a mechanism that acts to terminate periods of food intake at some point beyond satiation and prior to aversion, or the negative effects of extreme excess (nausea, bloating, etc.). The purpose of the present study was to assess the lateral habenula as a candidate region for such a mechanism, due to its connectivity to midbrain reward circuitry, sensitivity to metabolic signaling, and pronounced role in drug-related motivated behaviors. Two groups of male Sprague-Dawley rats were surgically implanted with bilateral guide cannula targeting the LHb. Rats were then habituated to feeding chambers, wherein locomotion and food intake were monitored throughout a two-hour session. One experimental group was tested in the presence of rat chow; the second group was instead given access to a sweetened fat diet. Each subject separately received a 0.2 μL vehicle (0.9% saline solution) and baclofen-muscimol (50 ng/0.2 μL of each drug dissolved in 0.9% saline) injection. Additionally, on a third injection day, each rat received an injection of mu-opioid agonist DAMGO (0.1 μg/0.2 μL) prior to placement in the chamber. LHb inactivation did not result in significant alterations in feeding behavior, but produced a consistent increase in locomotor activity in both experimental groups. Mu-opioid receptor stimulation increased feeding on standard chow, but decreased intake of the sweetened-fat diet. Although LHb inactivation did not increase feeding as predicted, the novel finding that mu opioid receptor stimulation decreased feeding on a highly palatable diet, but increased intake of rat chow, highlights a differential role for the LHb in regulating hedonic consummatory behavior.
Collapse
Affiliation(s)
| | | | - Wayne E Pratt
- Department of Psychology, Wake Forest University, USA.
| |
Collapse
|
18
|
Salaberry NL, Mendoza J. The circadian clock in the mouse habenula is set by catecholamines. Cell Tissue Res 2021; 387:261-274. [PMID: 34816282 DOI: 10.1007/s00441-021-03557-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
Circadian rhythms are those variations in behavioral and molecular processes of organisms that follow roughly 24 h cycles in the absence of any external cue. The hypothalamic suprachiasmatic nucleus (SCN) harbors the principal brain pacemaker driving circadian rhythms. The epithalamic habenula (Hb) contains a self-sustained circadian clock functionally coupled to the SCN. Anatomically, the Hb projects to the midbrain dopamine (DA) and serotonin (5-HT) systems, and it receives inputs from the forebrain, midbrain, and brainstem. The SCN is set by internal signals such as 5-HT or melatonin from the raphe nuclei and pineal gland, respectively. However, how the Hb clock is set by internal cues is not well characterized. Hence, in the present study, we determined whether DA, noradrenaline (NA), 5-HT, and the neuropeptides orexin (ORX) and vasopressin influence the Hb circadian clock. Using PER2::Luciferase transgenic mice, we found that the amplitude of the PER2 protein circadian oscillations from Hb explants was strongly affected by DA and NA. Importantly, these effects were dose-and region (rostral vs. caudal) dependent for NA, with a main effect in the caudal part of the Hb. Furthermore, ORX also induced a significant change in the amplitude of PER2 protein oscillations in the caudal Hb. In conclusion, catecholaminergic (DA, NA) and ORXergic transmission impacts the clock properties of the Hb clock likely contributing to the circadian regulation of motivated behaviors. Accordingly, pathological conditions that lead in alterations of catecholamine or ORX activity (drug intake, compulsive feeding) might affect the Hb clock and conduct to circadian disturbances.
Collapse
Affiliation(s)
- Nora L Salaberry
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 8 Allée du Général Rouvillois, Strasbourg, 67000, France
| | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 8 Allée du Général Rouvillois, Strasbourg, 67000, France.
| |
Collapse
|
19
|
Zhao S, Li R, Li H, Wang S, Zhang X, Wang D, Guo J, Li H, Li A, Tong T, Zhong H, Yang Q, Dong H. Lateral Hypothalamic Area Glutamatergic Neurons and Their Projections to the Lateral Habenula Modulate the Anesthetic Potency of Isoflurane in Mice. Neurosci Bull 2021; 37:934-946. [PMID: 33847915 PMCID: PMC8275739 DOI: 10.1007/s12264-021-00674-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/26/2020] [Indexed: 01/19/2023] Open
Abstract
The lateral hypothalamic area (LHA) plays a pivotal role in regulating consciousness transition, in which orexinergic neurons, GABAergic neurons, and melanin-concentrating hormone neurons are involved. Glutamatergic neurons have a large population in the LHA, but their anesthesia-related effect has not been explored. Here, we found that genetic ablation of LHA glutamatergic neurons shortened the induction time and prolonged the recovery time of isoflurane anesthesia in mice. In contrast, chemogenetic activation of LHA glutamatergic neurons increased the time to anesthesia and decreased the time to recovery. Optogenetic activation of LHA glutamatergic neurons during the maintenance of anesthesia reduced the burst suppression pattern of the electroencephalogram (EEG) and shifted EEG features to an arousal pattern. Photostimulation of LHA glutamatergic projections to the lateral habenula (LHb) also facilitated the emergence from anesthesia and the transition of anesthesia depth to a lighter level. Collectively, LHA glutamatergic neurons and their projections to the LHb regulate anesthetic potency and EEG features.
Collapse
Affiliation(s)
- Shiyi Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Rui Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Huiming Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Sa Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xinxin Zhang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Dan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Juan Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Huihui Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ao Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tingting Tong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Haixing Zhong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Qianzi Yang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
20
|
Metzger M, Souza R, Lima LB, Bueno D, Gonçalves L, Sego C, Donato J, Shammah-Lagnado SJ. Habenular connections with the dopaminergic and serotonergic system and their role in stress-related psychiatric disorders. Eur J Neurosci 2019; 53:65-88. [PMID: 31833616 DOI: 10.1111/ejn.14647] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/28/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
The habenula (Hb) is a phylogenetically old epithalamic structure differentiated into two nuclear complexes, the medial (MHb) and lateral habenula (LHb). After decades of search for a great unifying function, interest in the Hb resurged when it was demonstrated that LHb plays a major role in the encoding of aversive stimuli ranging from noxious stimuli to the loss of predicted rewards. Consistent with a role as an anti-reward center, aberrant LHb activity has now been identified as a key factor in the pathogenesis of major depressive disorder. Moreover, both MHb and LHb emerged as new players in the reward circuitry by primarily mediating the aversive properties of distinct drugs of abuse. Anatomically, the Hb serves as a bridge that links basal forebrain structures with monoaminergic nuclei in the mid- and hindbrain. So far, research on Hb has focused on the role of the LHb in regulating midbrain dopamine release. However, LHb/MHb are also interconnected with the dorsal (DR) and median (MnR) raphe nucleus. Hence, it is conceivable that some of the habenular functions are at least partly mediated by the complex network that links MHb/LHb with pontomesencephalic monoaminergic nuclei. Here, we summarize research about the topography and transmitter phenotype of the reciprocal connections between the LHb and ventral tegmental area-nigra complex, as well as those between the LHb and DR/MnR. Indirect MHb outputs via interpeduncular nucleus to state-setting neuromodulatory networks will also be commented. Finally, we discuss the role of specific LHb-VTA and LHb/MHb-raphe circuits in anxiety and depression.
Collapse
Affiliation(s)
- Martin Metzger
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Rudieri Souza
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Leandro B Lima
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Debora Bueno
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Luciano Gonçalves
- Department of Human Anatomy, Federal University of the Triângulo Mineiro, Uberaba, Brazil
| | - Chemutai Sego
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jose Donato
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sara J Shammah-Lagnado
- Department of Physiology & Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Godfrey N, Borgland SL. Diversity in the lateral hypothalamic input to the ventral tegmental area. Neuropharmacology 2019; 154:4-12. [DOI: 10.1016/j.neuropharm.2019.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/15/2019] [Accepted: 05/13/2019] [Indexed: 12/29/2022]
|
22
|
Cheon M, Park H, Rhim H, Chung C. Actions of Neuropeptide Y on Synaptic Transmission in the Lateral Habenula. Neuroscience 2019; 410:183-190. [PMID: 31082535 DOI: 10.1016/j.neuroscience.2019.04.053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/27/2019] [Accepted: 04/26/2019] [Indexed: 12/12/2022]
Abstract
Neuropeptide Y is a peptide neuromodulator with protective roles including anxiolytic and antidepressant-like effects in animal models of depression and post-traumatic stress disorder. The lateral habenula (LHb) is a brain region that encodes aversive information and is closely related with mood disorders. Although LHb neurons express NPY receptors, the physiological roles of NPY in this region remain uninvestigated. In this study, we examined the actions of NPY on synaptic transmission in the LHb using whole cell patch clamp recording. We observed that NPY inhibited excitatory neurotransmission in a subset of LHb neurons whereas potentiating in a small population of neurons. Inhibitory transmission remained unchanged by NPY application in a subset of neurons but was reduced in the majority of LHb neurons recorded. The overall outcome of NPY application was a decrease in the spontaneous firing rate of the LHb, leading to hypoactivation of the LHb. Our observations indicate that although NPY has divergent effects on excitatory and inhibitory transmission, NPY receptor activation decreases LHb activity, suggesting that the LHb may partly mediate the protective roles of NPY in the central nervous system.
Collapse
Affiliation(s)
- Myunghyun Cheon
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea
| | - Hyewon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 139-791, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul 05029, South Korea.
| |
Collapse
|
23
|
Animal Models of (or for) Aggression Reward, Addiction, and Relapse: Behavior and Circuits. J Neurosci 2019; 39:3996-4008. [PMID: 30833504 DOI: 10.1523/jneurosci.0151-19.2019] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/18/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Inappropriate and pathological aggression plays a leading role in the suffering and death of millions of people, and further places an untenable strain on the caregivers and families of those afflicted. In some cases, such as addictive drugs, aggression can be highly rewarding (appetitive) and continually pursued despite short- and long-term negative consequences. Similarly, recidivism (relapse) rates for repeat violent offenders are as high as relapse rates for drug addicts. Appetitive aggression and relapse to aggression seeking can be modeled in mice studies using conditioned place preference and self-administration procedures followed by a period of abstinence and subsequent tests for relapse to aggression preference and aggression seeking. These procedures allow for the study of the mechanisms that control the appetitive versus the consummatory (attack) phases of aggressive behavior. In this review, we first discuss the behavioral procedures developed to probe appetitive aggression in mouse models, spanning from Pavlovian to operant tasks, and we also describe the recently proposed phenomenon of "aggression addiction." Next, we discuss the pharmacological and circuit mechanisms of aggression conditioned place preference and aggression self-administration, seeking, and relapse, highlighting mechanistic congruence and divergence between appetitive and consummatory phases of aggression. We conclude by discussing clinical implications of the studies reviewed.
Collapse
|
24
|
Lateral parabrachial neurons innervate orexin neurons projecting to brainstem arousal areas in the rat. Sci Rep 2019; 9:2830. [PMID: 30808976 PMCID: PMC6391479 DOI: 10.1038/s41598-019-39063-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 01/16/2019] [Indexed: 01/31/2023] Open
Abstract
Orexin (ORX) neurons in the hypothalamus send their axons to arousal-promoting areas. We have previously shown that glutamatergic neurons in the lateral parabrachial nucleus (LPB) innervate ORX neurons. In this study, we examined potential pathways from the LPB to ORX neurons projecting to arousal-promoting areas in the brainstem by a combination of tract-tracing techniques in male Wistar rats. We injected the anterograde tracer biotinylated dextranamine (BDA) into the LPB and the retrograde tracer cholera toxin B subunit (CTb) into the ventral tegmental area, dorsal raphe nucleus, pedunculopontine tegmental nucleus, laterodorsal tegmental area, or locus coeruleus (LC). We then analyzed the BDA-labeled fibers and ORX-immunoreactive neurons in the hypothalamus. We found that double-labeled ORX and CTb neurons were the most abundant after CTb was injected into the LC. We also observed prominently overlapping distribution of BDA-labeled fibers, arising from neurons located in the lateral-most part of the dorsomedial nucleus and adjacent dorsal perifornical area. In these areas, we confirmed by confocal microscopy that BDA-labeled synaptophysin-immunoreactive axon terminals were in contiguity with cell bodies and dendrites of CTb-labeled ORX-immunoreactive neurons. These results suggest that the LPB innervates arousal-promoting areas via ORX neurons and is likely to promote arousal responses to stimuli.
Collapse
|
25
|
Noritake A, Nakamura K. Encoding prediction signals during appetitive and aversive Pavlovian conditioning in the primate lateral hypothalamus. J Neurophysiol 2019; 121:396-417. [DOI: 10.1152/jn.00247.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The lateral hypothalamus (LH), which plays a role in homeostatic functions such as appetite regulation, is also linked to arousal and motivational behavior. However, little is known about how these components are encoded in the LH. Thus cynomolgus monkeys were conditioned with two distinct contexts, i.e., an appetitive context with available rewards and an aversive context with predicted air puffs. Different LH neuron groups encoded different degrees of expectation, predictability, and risks of rewards in a specific manner. A nearly equal number of one-third of the recorded LH neurons showed a positive or negative correlation between their response to visual conditioned stimuli (CS) that predicted the probabilistic delivery of rewards (0%, 50%, and 100%) and the associative values. For another one-third of recorded neurons, a nearly equal number showed a positive or negative correlation between their responses to rewards [appetitive unconditioned stimulus (US)] and reward predictability. Some neurons exhibited their highest or lowest trace-period responses in the 50% reward trials. These response modulations were represented independently and overlaid on a consistent excitatory or inhibitory response across the conditioning events. LH neurons also showed consistent responses in the aversive context. However, the responses to aversive conditioning events depending on the air puff value and predictability were less common. The multifaceted modulation of consistent activity related to outcome predictions may reflect motivational and arousal signals. Furthermore, it may underlie the role the LH plays in the integration and relay of signals to cortices for adaptive and goal-directed physiological and behavioral responses to environmental changes. NEW & NOTEWORTHY The lateral hypothalamus (LH) is implicated in motivational and arousal behavior; however, the detailed information carried by single LH neurons remains unclear. We demonstrate that primate LH neurons encode multiple combinations of signals concerning different degrees of expectation, appreciation, and uncertainty of rewards in consistent responses across conditioning events and between different contexts. This multifaceted modulation of activity may underlie the role of the LH as a critical node integrating motivational signals with arousal signals.
Collapse
Affiliation(s)
- Atsushi Noritake
- Department of Physiology, Kansai Medical University, Hirakata-city, Osaka, Japan
- National Institute for Physiological Sciences, Okazaki-city, Aichi, Japan
| | - Kae Nakamura
- Department of Physiology, Kansai Medical University, Hirakata-city, Osaka, Japan
| |
Collapse
|
26
|
Abstract
Encoding and predicting aversive events are critical functions of circuits that support survival and emotional well-being. Maladaptive circuit changes in emotional valence processing can underlie the pathophysiology of affective disorders. The lateral habenula (LHb) has been linked to aversion and mood regulation through modulation of the dopamine and serotonin systems. We have defined the identity and function of glutamatergic (Vglut2) control of the LHb, comparing the role of inputs originating in the globus pallidus internal segment (GPi), and lateral hypothalamic area (LHA), respectively. We found that LHb-projecting LHA neurons, and not the proposed GABA/glutamate co-releasing GPi neurons, are responsible for encoding negative value. Monosynaptic rabies tracing of the presynaptic organization revealed a predominantly limbic input onto LHA Vglut2 neurons, while sensorimotor inputs were more prominent onto GABA/glutamate co-releasing GPi neurons. We further recorded the activity of LHA Vglut2 neurons, by imaging calcium dynamics in response to appetitive versus aversive events in conditioning paradigms. LHA Vglut2 neurons formed activity clusters representing distinct reward or aversion signals, including a population that responded to mild foot shocks and predicted aversive events. We found that the LHb-projecting LHA Vglut2 neurons encode negative valence and rapidly develop a prediction signal for negative events. These findings establish the glutamatergic LHA-LHb circuit as a critical node in value processing.
Collapse
|
27
|
A Human Polymorphism in CHRNA5 Is Linked to Relapse to Nicotine Seeking in Transgenic Rats. Curr Biol 2018; 28:3244-3253.e7. [PMID: 30293722 DOI: 10.1016/j.cub.2018.08.044] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/09/2018] [Accepted: 08/20/2018] [Indexed: 12/26/2022]
Abstract
Tobacco addiction is a chronic and relapsing disorder with an important genetic component that represents a major public health issue. Meta-analysis of large-scale human genome-wide association studies (GWASs) identified a frequent non-synonymous SNP in the gene coding for the α5 subunit of nicotinic acetylcholine receptors (α5SNP), which significantly increases the risk for tobacco dependence and delays smoking cessation. To dissect the neuronal mechanisms underlying the vulnerability to nicotine addiction in carriers of the α5SNP, we created rats expressing this polymorphism using zinc finger nuclease technology and evaluated their behavior under the intravenous nicotine-self-administration paradigm. The electrophysiological responses of their neurons to nicotine were also evaluated. α5SNP rats self-administered more nicotine at high doses and exhibited higher nicotine-induced reinstatement of nicotine seeking than wild-type rats. Higher reinstatement was associated with altered neuronal activity in several discrete areas that are interconnected, including in the interpeduncular nucleus (IPN), a GABAergic structure that strongly expresses α5-containing nicotinic receptors. The altered reactivity of IPN neurons of α5SNP rats to nicotine was confirmed electrophysiologically. In conclusion, the α5SNP polymorphism is a major risk factor for nicotine intake at high doses and for relapse to nicotine seeking in rats, a dual effect that reflects the human condition. Our results also suggest an important role for the IPN in the higher relapse to nicotine seeking observed in α5SNP rats.
Collapse
|
28
|
Salaberry NL, Hamm H, Felder-Schmittbuhl MP, Mendoza J. A suprachiasmatic-independent circadian clock(s) in the habenula is affected by Per gene mutations and housing light conditions in mice. Brain Struct Funct 2018; 224:19-31. [PMID: 30242505 DOI: 10.1007/s00429-018-1756-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/12/2018] [Indexed: 01/08/2023]
Abstract
For many years, the suprachiasmatic nucleus (SCN) was considered as the unique circadian pacemaker in the mammalian brain. Currently, it is known that other brain areas are able to oscillate in a circadian manner. However, many of them are dependent on, or synchronized by, the SCN. The Habenula (Hb), localized in the epithalamus, is a key nucleus for the regulation of monoamine activity (dopamine, serotonin) and presents circadian features; nonetheless, the clock properties of the Hb are not fully described. Here, we report, first, circadian expression of clock genes in the lateral habenula (LHb) under constant darkness (DD) condition in wild-type mice which is disturbed in double Per1-/--Per2Brdm1 clock-mutant mice. Second, using Per2::luciferase transgenic mice, we observed a self-sustained oscillatory ability (PER2::LUCIFERASE bioluminescence rhythmicity) in the rostral and caudal part of the Hb of arrhythmic SCN-ablated animals. Finally, in Per2::luciferase mice exposed to different lighting conditions (light-dark, constant darkness or constant light), the period or amplitude of PER2 oscillations, in both the rostral and caudal Hb, were similar. However, under DD condition or from SCN-lesioned mice, these two Hb regions were out of phase, suggesting an uncoupling of two putative Hb oscillators. Altogether, these results suggest that an autonomous clock in the rostral and caudal part of the Hb requires integrity of circadian genes to tick, and light information or SCN innervation to keep synchrony. The relevance of the Hb timing might reside in the regulation of circadian functions linked to motivational (reward) and emotional (mood) processes.
Collapse
Affiliation(s)
- Nora L Salaberry
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 5 rue Blaise Pascal, 67000, Strasbourg, France
| | - Hélène Hamm
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 5 rue Blaise Pascal, 67000, Strasbourg, France
| | | | - Jorge Mendoza
- Institute of Cellular and Integrative Neurosciences, CNRS UPR-3212, 5 rue Blaise Pascal, 67000, Strasbourg, France.
| |
Collapse
|
29
|
Namba MD, Tomek SE, Olive MF, Beckmann JS, Gipson CD. The Winding Road to Relapse: Forging a New Understanding of Cue-Induced Reinstatement Models and Their Associated Neural Mechanisms. Front Behav Neurosci 2018; 12:17. [PMID: 29479311 PMCID: PMC5811475 DOI: 10.3389/fnbeh.2018.00017] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
In drug addiction, cues previously associated with drug use can produce craving and frequently trigger the resumption of drug taking in individuals vulnerable to relapse. Environmental stimuli associated with drugs or natural reinforcers can become reliably conditioned to increase behavior that was previously reinforced. In preclinical models of addiction, these cues enhance both drug self-administration and reinstatement of drug seeking. In this review, we will dissociate the roles of conditioned stimuli as reinforcers from their modulatory or discriminative functions in producing drug-seeking behavior. As well, we will examine possible differences in neurobiological encoding underlying these functional differences. Specifically, we will discuss how models of drug addiction and relapse should more systematically evaluate these different types of stimuli to better understand the neurobiology underlying craving and relapse. In this way, behavioral and pharmacotherapeutic interventions may be better tailored to promote drug use cessation outcomes and long-term abstinence.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Seven E. Tomek
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - M. Foster Olive
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| | - Joshua S. Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, United States
| | - Cassandra D. Gipson
- Department of Psychology, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
30
|
Anatomical evidence for lateral hypothalamic innervation of the pontine A7 catecholamine cell group in rat. Neurosci Lett 2018; 668:80-85. [PMID: 29329908 DOI: 10.1016/j.neulet.2018.01.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/22/2017] [Accepted: 01/09/2018] [Indexed: 12/29/2022]
Abstract
Substantial behavioral evidence exists to support the idea that the lateral hypothalamus (LH) makes axonal connection with spinally-projecting noradrenergic neurons of the A7 catecholamine cell group in the pons. Through this putative projection, the LH modulates nociception via α1- and α2-adrenoceptors in the dorsal horn. We used double-label immunocytochemistry to demonstrate that axons from the LH labeled with the anterograde tracer biotinylated dextran amine (BDA) appose tyrosine hydroxylase-immunoreactive (TH-ir) neuron profiles in the A7 area. Other pontine areas labeled with BDA included the dorsomedial tegmental area, the pontine reticular nucleus, oral part, the caudal aspect of the dorsal raphe, the periaqueductal grey and the A6 area. To confirm the findings of the brightfield experiment, we used confocal microscopy to identify axons from the LH labeled with the anterograde tracer Fluoro-Ruby co-localized with TH-ir dendrites and cell bodies in the A7 cell group. These findings provide an anatomical substrate for behavioral studies in which stimulation of the LH modifies nociception in the spinal cord via norepinephrine.
Collapse
|
31
|
Zahm DS, Root DH. Review of the cytology and connections of the lateral habenula, an avatar of adaptive behaving. Pharmacol Biochem Behav 2017; 162:3-21. [PMID: 28647565 PMCID: PMC5659881 DOI: 10.1016/j.pbb.2017.06.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 05/02/2017] [Accepted: 06/13/2017] [Indexed: 12/21/2022]
Abstract
The cytology and connections of the lateral habenula (LHb) are reviewed. The habenula is first introduced, after which the cytology of the LHb is discussed mainly with reference to cell types, general topography and descriptions of subnuclei. An overview of LHb afferent connections is given followed by some details about the projections to LHb from a number of structures. An overview of lateral habenula efferent connections is given followed by some details about the projections from LHb to a number of structures. In considering the afferent and efferent connections of the LHb some attention is given to the relative validity of regarding it as a bi-partite structure featuring 'limbic' and 'pallidal' parts. The paper ends with some concluding remarks about the relative place of the LHb in adaptive behaving.
Collapse
Affiliation(s)
- Daniel S Zahm
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S. Grand Blvd., Saint Louis, MO 63104, United States.
| | - David H Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO 80309, United States.
| |
Collapse
|
32
|
Mendoza J. Circadian neurons in the lateral habenula: Clocking motivated behaviors. Pharmacol Biochem Behav 2017; 162:55-61. [PMID: 28666896 DOI: 10.1016/j.pbb.2017.06.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/30/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022]
Abstract
The main circadian clock in mammals is located in the hypothalamic suprachiasmatic nucleus (SCN), however, central timing mechanisms are also present in other brain structures beyond the SCN. The lateral habenula (LHb), known for its important role in the regulation of the monoaminergic system, contains such a circadian clock whose molecular and cellular mechanisms as well as functional role are not well known. However, since monoaminergic systems show circadian activity, it is possible that the LHb-clock's role is to modulate the rhythmic activity of the dopamine, serotonin and norephinephrine systems, and associated behaviors. Moreover, the LHb is involved in different pathological states such as depression, addiction and schizophrenia, states in which sleep and circadian alterations have been reported. Thus, perturbations of circadian activity in the LHb might, in part, be a cause of these rhythmic alterations in psychiatric ailments. In this review the current state of the LHb clock and its possible implications in the control of monoaminergic systems rhythms, motivated behaviors (e.g., feeding, drug intake) and depression (with circadian disruptions and altered motivation) will be discussed.
Collapse
Affiliation(s)
- Jorge Mendoza
- Institute of Cellular and Integrative Neuroscience, CNRS-UPR 3212 Strasbourg France, 5 rue Blaise Pascal, 67084 cedex Strasbourg, France.
| |
Collapse
|
33
|
Sheth C, Furlong TM, Keefe KA, Taha SA. The lateral hypothalamus to lateral habenula projection, but not the ventral pallidum to lateral habenula projection, regulates voluntary ethanol consumption. Behav Brain Res 2017; 328:195-208. [PMID: 28432009 PMCID: PMC5500222 DOI: 10.1016/j.bbr.2017.04.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/23/2017] [Accepted: 04/17/2017] [Indexed: 11/26/2022]
Abstract
The lateral habenula (LHb) is an epithalamic brain region implicated in aversive processing via negative modulation of midbrain dopamine (DA) and serotonin (5-HT) systems. Given the role of the LHb in inhibiting DA and 5-HT systems, it is thought to be involved in various psychiatric pathologies, including drug addiction. In support, it has been shown that LHb plays a critical role in cocaine- and ethanol-related behaviors, most likely by mediating drug-induced aversive conditioning. In our previous work, we showed that LHb lesions increased voluntary ethanol consumption and operant ethanol self-administration and blocked yohimbine-induced reinstatement of ethanol self-administration. LHb lesions also attenuated ethanol-induced conditioned taste aversion suggesting that a mechanism for the increased intake of ethanol may be reduced aversion learning. However, whether afferents to the LHb are required for mediating effects of the LHb on these behaviors remained to be investigated. Our present results show that lesioning the fiber bundle carrying afferent inputs to the LHb, the stria medullaris (SM), increases voluntary ethanol consumption, suggesting that afferent structures projecting to the LHb are important for mediating ethanol-directed behaviors. We then chose two afferent structures as the focus of our investigation. We specifically studied the role of the inputs from the lateral hypothalamus (LH) and ventral pallidum (VP) to the LHb in ethanol-directed behaviors. Our results show that the LH-LHb projection is necessary for regulating voluntary ethanol consumption. These results are an important first step towards understanding the functional role of afferents to LHb with regard to ethanol consumption.
Collapse
Affiliation(s)
- Chandni Sheth
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112-5820, USA.
| | - Teri M Furlong
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112-5820, USA
| | - Kristen A Keefe
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112-5820, USA
| | - Sharif A Taha
- Department of Pharmacology and Toxicology, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112-5820, USA
| |
Collapse
|
34
|
Abstract
Over the past 20years, substantive research has firmly implicated the lateral habenula in myriad neural processes including addiction, depression, and sleep. More recently, evidence has emerged suggesting that the lateral habenula is a component of the brain's intrinsic daily or circadian timekeeping system. This system centers on the master circadian pacemaker in the suprachiasmatic nuclei of the hypothalamus that is synchronized to the external world through environmental light information received directly from the eye. Rhythmic clock gene expression in suprachiasmatic neurons drives variation in their electrical activity enabling communication of temporal information, and the organization of circadian rhythms in downstream targets. Here, we review the evidence implicating the lateral habenula as part of an extended neural circadian system. We consider findings suggesting that the lateral habenula is a recipient of circadian signals from the suprachiasmatic nuclei as well as light information from the eye. Further we examine the proposition that the lateral habenula itself expresses intrinsic clock gene and neuronal rhythms. We then speculate on how circadian information communicated from the lateral habenula could influence activity and function in downstream targets such as the ventral tegmental area and raphe nuclei.
Collapse
Affiliation(s)
| | - Hugh D Piggins
- Faculty of Biology, Medicine and Health, University of Manchester, M13 9PT, UK.
| |
Collapse
|
35
|
Batalla A, Homberg JR, Lipina TV, Sescousse G, Luijten M, Ivanova SA, Schellekens AFA, Loonen AJM. The role of the habenula in the transition from reward to misery in substance use and mood disorders. Neurosci Biobehav Rev 2017; 80:276-285. [PMID: 28576510 DOI: 10.1016/j.neubiorev.2017.03.019] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/14/2017] [Indexed: 12/18/2022]
Abstract
The habenula (Hb) is an evolutionary well-conserved structure located in the epithalamus. The Hb receives inputs from the septum, basal ganglia, hypothalamus, anterior cingulate and medial prefrontal cortex, and projects to several midbrain centers, most importantly the inhibitory rostromedial tegmental nucleus (RMTg) and the excitatory interpeduncular nucleus (IPN), which regulate the activity of midbrain monoaminergic nuclei. The Hb is postulated to play a key role in reward and aversion processing across species, including humans, and to be implicated in the different stages of transition from recreational drug intake to addiction and co-morbid mood disorders. The Hb is divided into two anatomically and functionally distinct nuclei, the lateral (LHb) and the medial (MHb), which are primarily involved in reward-seeking (LHb) and misery-fleeing (MHb) behavior by controlling the RMTg and IPN, respectively. This review provides a neuroanatomical description of the Hb, discusses preclinical and human findings regarding its role in the development of addiction and co-morbid mood disorders, and addresses future directions in this area.
Collapse
Affiliation(s)
- Albert Batalla
- Radboud University Medical Center, Department of Psychiatry, Reinier Postlaan 10, 6500 HB, Nijmegen, The Netherlands; Radboud University, Nijmegen Institute for Scientist-Practitioners in Addiction, Toernooiveld 5, 6525 ED, Nijmegen, The Netherlands.
| | - Judith R Homberg
- Radboud University Medical Center, Department of Cognitive Neuroscience, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Tatiana V Lipina
- Federal State Budgetary Scientific Institution, Scientific Research Institute of Physiology and Basic Medicine, Timakova 4, 630117, Novosibirsk, Russia; Novosibirsk State University, Pirogova 2, 630090, Novosibirsk, Russia.
| | - Guillaume Sescousse
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands.
| | - Maartje Luijten
- Behavioural Science Institute, Radboud University, Montessorilaan 3, 6525 HR, Nijmegen, The Netherlands.
| | - Svetlana A Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, Aleutskaya street 4, 634014, Tomsk, Russian Federation; National Research Tomsk Polytechnic University, Lenin Avenue, 30, 634050, Tomsk, Russian Federation.
| | - Arnt F A Schellekens
- Radboud University Medical Center, Department of Psychiatry, Reinier Postlaan 10, 6500 HB, Nijmegen, The Netherlands; Radboud University, Nijmegen Institute for Scientist-Practitioners in Addiction, Toernooiveld 5, 6525 ED, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition and Behaviour, Radboud University, Kapittelweg 29, 6525 EN, Nijmegen, The Netherlands.
| | - Anton J M Loonen
- Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713AV, Groningen, The Netherlands; GGZ Westelijk Noord-Brabant, Hoofdlaan 8, 4661AA, Halsteren, The Netherlands.
| |
Collapse
|
36
|
Flanigan M, Aleyasin H, Takahashi A, Golden SA, Russo SJ. An emerging role for the lateral habenula in aggressive behavior. Pharmacol Biochem Behav 2017; 162:79-86. [PMID: 28499809 DOI: 10.1016/j.pbb.2017.05.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/24/2017] [Accepted: 05/05/2017] [Indexed: 12/19/2022]
Abstract
Inter-male aggression is an essential component of social behavior in organisms from insects to humans. However, when expressed inappropriately, aggression poses significant threats to the mental and physical health of both the aggressor and the target. Inappropriate aggression is a common feature of numerous neuropsychiatric disorders in humans and has been hypothesized to result from the atypical activation of reward circuitry in response to social targets. The lateral habenula (LHb) has recently been identified as a major node of the classical reward circuitry and inhibits the release of dopamine from the midbrain to signal negative valence. Here, we discuss the evidence linking LHb function to aggression and its valence, arguing that strong LHb outputs to the ventral tegmental area (VTA) and the dorsal raphe nucleus (DRN) are likely to play roles in aggression and its rewarding components. Future studies should aim to elucidate how various inputs and outputs of the LHb shape motivation and reward in the context of aggression.
Collapse
Affiliation(s)
- Meghan Flanigan
- Fishberg Department of Neuroscience and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hossein Aleyasin
- Fishberg Department of Neuroscience and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aki Takahashi
- Fishberg Department of Neuroscience and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; University of Tsukuba, Tsukuba, Japan
| | - Sam A Golden
- National Institute of Drug Abuse, Baltimore, MD, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
37
|
Petzel A, Bernard R, Poller WC, Veh RW. Anterior and posterior parts of the rat ventral tegmental area and the rostromedial tegmental nucleus receive topographically distinct afferents from the lateral habenular complex. J Comp Neurol 2017; 525:2310-2327. [DOI: 10.1002/cne.24200] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 02/14/2017] [Accepted: 02/17/2017] [Indexed: 02/03/2023]
Affiliation(s)
- Anja Petzel
- Charité - Universitätsmedizin Berlin, Institut für Zell- und Neurobiologie; Berlin Germany
| | - René Bernard
- Charité - Universitätsmedizin Berlin, Institut für Zell- und Neurobiologie; Berlin Germany
| | - Wolfram C. Poller
- Charité - Universitätsmedizin Berlin, Institut für Zell- und Neurobiologie; Berlin Germany
| | - Rüdiger W. Veh
- Charité - Universitätsmedizin Berlin, Institut für Zell- und Neurobiologie; Berlin Germany
| |
Collapse
|
38
|
Abstract
The neural control of appetite is important for understanding motivated behavior as well as the present rising prevalence of obesity. Over the past several years, new tools for cell type-specific neuron activity monitoring and perturbation have enabled increasingly detailed analyses of the mechanisms underlying appetite-control systems. Three major neural circuits strongly and acutely influence appetite but with notably different characteristics. Although these circuits interact, they have distinct properties and thus appear to contribute to separate but interlinked processes influencing appetite, thereby forming three pillars of appetite control. Here, we summarize some of the key characteristics of appetite circuits that are emerging from recent work and synthesize the findings into a provisional framework that can guide future studies.
Collapse
Affiliation(s)
- Scott M Sternson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147;
| | - Anne-Kathrin Eiselt
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia 20147;
| |
Collapse
|
39
|
Loonen AJM, Ivanova SA. Circuits Regulating Pleasure and Happiness: The Evolution of the Amygdalar-Hippocampal-Habenular Connectivity in Vertebrates. Front Neurosci 2016; 10:539. [PMID: 27920666 PMCID: PMC5118621 DOI: 10.3389/fnins.2016.00539] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Accepted: 11/04/2016] [Indexed: 01/05/2023] Open
Abstract
Appetitive-searching (reward-seeking) and distress-avoiding (misery-fleeing) behavior are essential for all free moving animals to stay alive and to have offspring. Therefore, even the oldest ocean-dwelling animal creatures, living about 560 million years ago and human ancestors, must have been capable of generating these behaviors. The current article describes the evolution of the forebrain with special reference to the development of the misery-fleeing system. Although, the earliest vertebrate ancestor already possessed a dorsal pallium, which corresponds to the human neocortex, the structure and function of the neocortex was acquired quite recently within the mammalian evolutionary line. Up to, and including, amphibians, the dorsal pallium can be considered to be an extension of the medial pallium, which later develops into the hippocampus. The ventral and lateral pallium largely go up into the corticoid part of the amygdala. The striatopallidum of these early vertebrates becomes extended amygdala, consisting of centromedial amygdala (striatum) connected with the bed nucleus of the stria terminalis (pallidum). This amygdaloid system gives output to hypothalamus and brainstem, but also a connection with the cerebral cortex exists, which in part was created after the development of the more recent cerebral neocortex. Apart from bidirectional connectivity with the hippocampal complex, this route can also be considered to be an output channel as the fornix connects the hippocampus with the medial septum, which is the most important input structure of the medial habenula. The medial habenula regulates the activity of midbrain structures adjusting the intensity of the misery-fleeing response. Within the bed nucleus of the stria terminalis the human homolog of the ancient lateral habenula-projecting globus pallidus may exist; this structure is important for the evaluation of efficacy of the reward-seeking response. The described organization offers a framework for the regulation of the stress response, including the medial habenula and the subgenual cingulate cortex, in which dysfunction may explain the major symptoms of mood and anxiety disorders.
Collapse
Affiliation(s)
- Anton J. M. Loonen
- Department of Pharmacy, University of GroningenGroningen, Netherlands
- GGZ Westelijk Noord-Brabant (GGZ-WNB)Halsteren, Netherlands
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of SciencesTomsk, Russia
- Department of Ecology and Basic Safety, National Research Tomsk Polytechnic UniversityTomsk, Russia
| |
Collapse
|
40
|
Fakhoury M, Voyer D, Lévesque D, Rompré PP. Effect of electrolytic lesions of the dorsal diencephalic conduction system on the distribution of Fos-like immunoreactivity induced by rewarding electrical stimulation. Neuroscience 2016; 334:214-225. [DOI: 10.1016/j.neuroscience.2016.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/27/2016] [Accepted: 08/02/2016] [Indexed: 11/30/2022]
|
41
|
Salaberry NL, Mateo M, Mendoza J. The Clock Gene Rev-Erbα Regulates Methamphetamine Actions on Circadian Timekeeping in the Mouse Brain. Mol Neurobiol 2016; 54:5327-5334. [PMID: 27581301 DOI: 10.1007/s12035-016-0076-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/23/2016] [Indexed: 12/21/2022]
Abstract
Circadian rhythms are strongly affected by drugs. In rodents, chronic methamphetamine (METH) intake changes circadian activity rhythms, mainly by altering light synchronization that generates the expression of a free-running rhythm with a period longer than 24 h and a second behavioral component that is independent of the main suprachiasmatic (SCN) clock. Although a number of clock genes do not appear to be involved in the effects of METH on circadian behavior, the molecular clockwork controlling these changes is still unclear. Therefore, we investigated the role of the clock gene Rev-Erbα in METH-induced behavioral and molecular responses using knockout mice and their wild-type littermates. Chronic intake of METH alters period circadian behavior of wild-type mice. However, in mice lacking the clock gene Rev-Erbα METH had no effect on their behavioral rhythms. Furthermore, PER2 bioluminescence rhythms in two extra-SCN brain oscillators, the dorsomedial hypothalamus and the habenula, were altered by METH in wild type but not in KO mice. Together, the present results implicate Rev-Erbα in the modulation of the circadian responses to METH and may provide a better comprehension into the mechanisms underlying circadian alterations provoked by drug addiction.
Collapse
Affiliation(s)
- Nora L Salaberry
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France
| | - Maria Mateo
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France
| | - Jorge Mendoza
- CNRS UPR-3212, Institute of Cellular and Integrative Neurosciences, 5 rue Blaise Pascal, 67084, Strasbourg, Cedex, France.
| |
Collapse
|
42
|
Margolis EB, Fields HL. Mu Opioid Receptor Actions in the Lateral Habenula. PLoS One 2016; 11:e0159097. [PMID: 27427945 PMCID: PMC4948872 DOI: 10.1371/journal.pone.0159097] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/27/2016] [Indexed: 11/19/2022] Open
Abstract
Increased activity of lateral habenula (LHb) neurons is correlated with aversive states including pain, opioid abstinence, rodent models of depression, and failure to receive a predicted reward. Agonists at the mu opioid receptor (MOR) are among the most powerful rewarding and pain relieving drugs. Injection of the MOR agonist morphine directly into the habenula produces analgesia, raising the possibility that MOR acts locally within the LHb. Consequently, we examined the synaptic actions of MOR agonists in the LHb using whole cell patch clamp recording. We found that the MOR selective agonist DAMGO inhibits a subset of LHb neurons both directly and by inhibiting glutamate release onto these cells. Paradoxically, DAMGO also presynaptically inhibited GABA release onto most LHb neurons. The behavioral effect of MOR activation will thus depend upon both the level of intrinsic neuronal activity in the LHb and the balance of activity in glutamate and GABA inputs to different LHb neuronal populations.
Collapse
Affiliation(s)
- Elyssa B. Margolis
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| | - Howard L. Fields
- Department of Neurology, The Wheeler Center for the Neurobiology of Addiction, Alcoholism and Addiction Research Group, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
43
|
Lateral Hypothalamic Area Glutamatergic Neurons and Their Projections to the Lateral Habenula Regulate Feeding and Reward. J Neurosci 2016; 36:302-11. [PMID: 26758824 DOI: 10.1523/jneurosci.1202-15.2016] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED The overconsumption of calorically dense, highly palatable foods is thought to be a major contributor to the worldwide obesity epidemic; however, the precise neural circuits that directly regulate hedonic feeding remain elusive. Here, we show that lateral hypothalamic area (LHA) glutamatergic neurons, and their projections to the lateral habenula (LHb), negatively regulate the consumption of palatable food. Genetic ablation of LHA glutamatergic neurons increased daily caloric intake and produced weight gain in mice that had access to a high-fat diet, while not altering general locomotor activity. Anterior LHA glutamatergic neurons send a functional glutamatergic projection to the LHb, a brain region involved in processing aversive stimuli and negative reward prediction outcomes. Pathway-specific, optogenetic stimulation of glutamatergic LHA-LHb circuit resulted in detectable glutamate-mediated EPSCs as well as GABA-mediated IPSCs, although the net effect of neurotransmitter release was to increase the firing of most LHb neurons. In vivo optogenetic inhibition of LHA-LHb glutamatergic fibers produced a real-time place preference, whereas optogenetic stimulation of LHA-LHb glutamatergic fibers had the opposite effect. Furthermore, optogenetic inhibition of LHA-LHb glutamatergic fibers acutely increased the consumption of a palatable liquid caloric reward. Collectively, these results demonstrate that LHA glutamatergic neurons are well situated to bidirectionally regulate feeding and potentially other behavioral states via their functional circuit connectivity with the LHb and potentially other brain regions. SIGNIFICANCE STATEMENT In this study, we show that the genetic ablation of LHA glutamatergic neurons enhances caloric intake. Some of these LHA glutamatergic neurons project to the lateral habenula, a brain area important for generating behavioral avoidance. Optogenetic stimulation of this circuit has net excitatory effects on postsynaptic LHb neurons. This is the first study to characterize the functional connectivity and behavioral relevance of this circuit within the context of feeding and reward-related behavior.
Collapse
|
44
|
Evely KM, Hudson RL, Dubocovich ML, Haj-Dahmane S. Melatonin receptor activation increases glutamatergic synaptic transmission in the rat medial lateral habenula. Synapse 2016; 70:181-186. [PMID: 26799638 DOI: 10.1002/syn.21892] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Melatonin (MLT) is secreted from the pineal gland and mediates its physiological effects through activation of two G protein-coupled receptors, MT1 and MT2 . These receptors are expressed in several brain areas, including the habenular complex, a pair of nuclei that relay information from forebrain to midbrain and modulate a plethora of behaviors, including sleep, mood, and pain. However, so far, the precise mechanisms by which MLT control the function of habenula neurons remain unknown. Using whole cell recordings from male rat brain slices, we examined the effects of MLT on the excitability of medial lateral habenula (MLHb) neurons. We found that MLT had no significant effects on the intrinsic excitability of MLHb neurons, but profoundly increased the amplitude of glutamate-mediated evoked excitatory post-synaptic currents (EPSC). The increase in strength of glutamate synapses onto MLHb neurons was mediated by an increase in glutamate release. The MLT-induced increase in glutamatergic synaptic transmission was blocked by the competitive MT1 /MT2 receptor antagonist luzindole (LUZ). These results unravel a potential cellular mechanism by which MLT receptor activation enhances the excitability of MLHb neurons. The MLT-mediated control of glutamatergic inputs to the MLHb may play a key role in the modulation of various behaviors controlled by the habenular complex.
Collapse
Affiliation(s)
- Katherine M Evely
- Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, New York
- Research Institute on Addictions, University at Buffalo, State University of New York, Buffalo, New York
| | - Randall L Hudson
- Department of Physiology and Biophysics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York
| | - Margarita L Dubocovich
- Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, New York
| | - Samir Haj-Dahmane
- Department of Pharmacology and Toxicology, University at Buffalo, State University of New York, Buffalo, New York
- Research Institute on Addictions, University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
45
|
Microarray analysis of transcripts with elevated expressions in the rat medial or lateral habenula suggest fast GABAergic excitation in the medial habenula and habenular involvement in the regulation of feeding and energy balance. Brain Struct Funct 2016; 221:4663-4689. [DOI: 10.1007/s00429-016-1195-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 01/29/2016] [Indexed: 10/22/2022]
|
46
|
Stuber GD, Wise RA. Lateral hypothalamic circuits for feeding and reward. Nat Neurosci 2016; 19:198-205. [PMID: 26814589 PMCID: PMC4927193 DOI: 10.1038/nn.4220] [Citation(s) in RCA: 328] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022]
Abstract
In experiments conducted over 60 years ago, the lateral hypothalamic area (LHA) was identified as a critical neuroanatomical substrate for motivated behavior. Electrical stimulation of the LHA induces voracious feeding even in well-fed animals. In the absence of food, animals will work tirelessly, often lever-pressing thousands of times per hour, for electrical stimulation at the same site that provokes feeding, drinking and other species-typical motivated behaviors. Here we review the classic findings from electrical stimulation studies and integrate them with more recent work that has used contemporary circuit-based approaches to study the LHA. We identify specific anatomically and molecularly defined LHA elements that integrate diverse information arising from cortical, extended amygdala and basal forebrain networks to ultimately generate a highly specified and invigorated behavioral state conveyed via LHA projections to downstream reward and feeding-specific circuits.
Collapse
Affiliation(s)
- Garret D. Stuber
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Roy A. Wise
- Intramural Research Program National Institute on Drug Abuse, NIH/DHHS, Baltimore, MD 21224, USA
| |
Collapse
|
47
|
A comparison of a behavioral weight loss program to a stress management program: A pilot randomized controlled trial. Nutrition 2016; 32:904-9. [PMID: 27138110 DOI: 10.1016/j.nut.2016.01.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/07/2016] [Accepted: 01/09/2016] [Indexed: 12/25/2022]
Abstract
OBJECTIVES This study compared a behavioral weight loss program (BWL) with a stress management-based program, Emotional Brain Training (EBT), on weight loss, blood pressure, depression, perceived stress, diet, and physical activity. METHODS Subjects with a body mass index (BMI) of >28 and <45 kg/m(2) were recruited in Lexington, Kentucky in January 2014 and randomized to BWL or EBT for a 20-week intervention. Of those recruited, 49 participants were randomized to EBT or BWL. Randomization and allocation to group were performed using SPSS software. Weight, blood pressure, depression, perceived stress, dietary intake, and physical activity were measured at baseline, 10 week, and 20 week. Linear models for change over time were fit to calculate 95% confidence intervals of intervention effects. RESULTS BWL produced greater changes in BMI than EBT at both 10 (P = 0.02) and 20 wk (P = 0.03). At 10 wk, both EBT and BWL improved BMI, systolic blood pressure, depression and perceived stress (P < 0.05). BWL also improved diastolic blood pressure (P = 0.005). At 20 wk, EBT maintained improvements in BMI, systolic blood pressure, depression, and perceived stress while BWL maintained improvements only in BMI and depression (P < 0.05). CONCLUSIONS BWL produced greater weight loss than EBT; however, EBT produced sustained improvements in stress, depression, and systolic blood pressure. A combination of the two approaches should be explored.
Collapse
|
48
|
Role of the dorsal diencephalic conduction system in the brain reward circuitry. Behav Brain Res 2015; 296:431-441. [PMID: 26515931 DOI: 10.1016/j.bbr.2015.10.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 12/13/2022]
Abstract
Previous work with psychophysically based studies suggests that electrolytic lesion of the habenula, which lies in the dorsal diencephalic conduction system (DDC), degrades the intracranial self-stimulation (ICSS). This experiment was aimed at studying the importance of the DDC in brain stimulation reward, and its connections with other areas that support operant responding for brain stimulation. For this purpose, rats were implanted with stimulating electrodes at the dorsal raphe (DR) and lateral hypothalamus (LH), and lesioning electrodes in the medial forebrain bundle (MFB) and the DDC. Rats were trained to self-administer the stimulation at three different current intensities and were tested daily for changes in reward thresholds, defined as the pulse frequency required for half-maximal responding. The lesions were done at the DDC and the MFB, and were separated by two weeks interval during which the rats were tested for self-stimulation. At the end of the experiment, rats were transcardially perfused and their brains collected to determine the extent of the lesions and the locations of the stimulation sites. Results show that lesions at both the DDC and MFB produce larger and longer-lasting increases in the reward thresholds (upto 0.40 log10 units) than lesions at either pathway alone (upto 0.25 log10 units), and were more effective in attenuating the reward induced by the LH stimulation. These results suggest that there exist two parallel pathways, the MFB and the DDC, which could constitute a viable route for the reward signal triggered by ICSS.
Collapse
|
49
|
Lecca S, Meye FJ, Mameli M. The lateral habenula in addiction and depression: an anatomical, synaptic and behavioral overview. Eur J Neurosci 2014; 39:1170-8. [PMID: 24712996 DOI: 10.1111/ejn.12480] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/10/2013] [Accepted: 12/12/2013] [Indexed: 02/06/2023]
Abstract
The lateral habenula (LHb) is an epithalamic region with a crucial role in the regulation of midbrain monoaminergic systems. Over the past few years a renewed interest in the LHb has emerged due to studies highlighting its central role in encoding rewarding and aversive aspects of stimuli. Moreover, an increasing number of functional as well as behavioral indications provide substantial evidence supporting a role of LHb in neuropsychiatric diseases, including mood disorders and drug addiction. Cellular and synaptic adaptations in the LHb may therefore represent a critical phenomenon in the etiology of these diseases. In the current review we describe the anatomical and functional connections allowing the LHb to control the dopamine and serotonin systems, as well as possible roles of these connections in motivated behaviors and neuropsychiatric disorders. Finally, we discuss how drug exposure and stressful conditions alter the cellular physiology of the LHb, highlighting a role for the LHb in the context of drug addiction and depression.
Collapse
Affiliation(s)
- Salvatore Lecca
- Institut du Fer à Moulin, 75005, Paris, France; Inserm, UMR-S 839, 75005, Paris, France; Université Pierre et Marie Curie, 75005, Paris, France
| | | | | |
Collapse
|
50
|
Wheeler DS, Wan S, Miller A, Angeli N, Adileh B, Hu W, Holland PC. Role of lateral hypothalamus in two aspects of attention in associative learning. Eur J Neurosci 2014; 40:2359-77. [PMID: 24750426 PMCID: PMC4641454 DOI: 10.1111/ejn.12592] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/12/2014] [Accepted: 03/17/2014] [Indexed: 11/28/2022]
Abstract
Orexin (hypocretin) and melanin-concentrating hormone (MCH) neurons are unique to the lateral hypothalamic (LH) region, but project throughout the brain. These cell groups have been implicated in a variety of functions, including reward learning, responses to stimulants, and the modulation of attention, arousal and the sleep/wakefulness cycle. Here, we examined roles for LH in two aspects of attention in associative learning shown previously to depend on intact function in major targets of orexin and MCH neurons. In experiments 1 and 2, unilateral orexin-saporin lesions of LH impaired the acquisition of conditioned orienting responses (ORs) and bilaterally suppressed FOS expression in the amygdala central nucleus (CeA) normally observed in response to food cues that provoke conditioned ORs. Those cues also induced greater FOS expression than control cues in LH orexin neurons, but not in MCH neurons. In experiment 3, unilateral orexin-saporin lesions of LH eliminated the cue associability enhancements normally produced by the surprising omission of an expected event. The magnitude of that impairment was positively correlated with the amount of LH damage and with the loss of orexin neurons in particular, but not with the loss of MCH neurons. We suggest that the effects of the LH orexin-saporin lesions were mediated by their effect on information processing in the CeA, known to be critical to both behavioral phenomena examined here. The results imply close relations between LH motivational amplification functions and attention, and may inform our understanding of disorders in which motivational and attentional impairments co-occur.
Collapse
Affiliation(s)
- Daniel S Wheeler
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, 21218, USA
| | | | | | | | | | | | | |
Collapse
|