1
|
Li J, Wei Y, Xiang J, Zhang D. Role of the ventral tegmental area in general anesthesia. Eur J Pharmacol 2025; 986:177145. [PMID: 39566814 DOI: 10.1016/j.ejphar.2024.177145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/22/2024]
Abstract
The ventral tegmental area (VTA), located in the midbrain, plays a pivotal role in the regulation of many important behaviors, such as reward, addiction, aversion, memory, learning, and sleep-wakefulness cycles. The majority of VTA neurons are dopaminergic neurons, although there is a significant proportion of GABAergic neurons and few glutamatergic neurons. These neuronal types project to different brain regions, thus mediating various biological functions. Therefore, the diverse roles of the VTA might depend on its heterogeneous neuronal types and projecting circuits. General anesthesia and sleep-wakefulness cycles share the feature of reversible loss of consciousness, and several common neural mechanisms underlie these two conditions. In addition to the well-known regulatory role of VTA in sleep-wakefulness, emerging evidence has demonstrated that VTA activity is also associated with promoting emergence from general anesthesia. Herein, we reviewed the literature and summarized the evidence regarding the modulation of the VTA by general anesthesia in rodents, which will improve the understanding of the modulatory mechanism of the VTA in general anesthesia.
Collapse
Affiliation(s)
- Jia Li
- Department of Anesthesiology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710000, China; Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China.
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, 518100, China
| | - Jiaxin Xiang
- Department of Anesthesiology, Weill Cornell Medicine, New York, 10065, USA
| | - Donghang Zhang
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, 430048, China; Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
2
|
Castro-Vildosola J, Bryan CA, Tajamal N, Jonnalagadda SA, Kasturi A, Tilly J, Garcia I, Kumar R, Fried NT, Hala T, Corbett BF. Sphingosine-1-phosphate receptor 3 activation promotes sociability and regulates transcripts important for anxiolytic-like behavior. Brain Behav Immun 2024; 124:205-217. [PMID: 39638159 DOI: 10.1016/j.bbi.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024] Open
Abstract
We previously demonstrated that sphingosine-1-phosphate receptor 3 (S1PR3) in the medial prefrontal cortex (mPFC) prevents reductions in sociability normally caused by stress. S1PR3 is a ubiquitously expressed G-protein coupled receptor that regulates immune system function, although its regulation of other biological processes is not well understood. Pharmacological activators of S1PR3 might provide important insights for understanding the neural substrates underlying sociability. Here we show that in mice, systemic injections of an S1PR3-specific agonist, CYM5541, promotes sociability in males and females whereas an S1PR3-specific antagonist, CAY10444, increases amygdala activation and increases social avoidance, particularly in females. S1PR3 expression is increased in the mPFC and dentate gyrus of females compared to males. RNA sequencing in the mPFC reveals that S1PR3 activation alters the expression of transcripts related to immune function, neurotransmission, transmembrane ion transport, and intracellular signaling. This work provides evidence that S1PR3 agonists, which have classically been used as immune modulators, might also be used to promote social behavior and, potentially, relieve symptoms of social anxiety. S1PR3 might be an important hub gene for mitigating maladaptive effects of stress as it reduces inflammatory processes, increases transcripts linked to anxiolytic neurotransmission, and promotes social behavior.
Collapse
Affiliation(s)
| | - Chris-Ann Bryan
- Department of Biology, Rutgers University, Camden, NJ, USA; Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nasira Tajamal
- Department of Biology, Rutgers University, Camden, NJ, USA
| | | | - Akhila Kasturi
- Department of Biology, Rutgers University, Camden, NJ, USA
| | | | - Isabel Garcia
- Department of Biology, Rutgers University, Camden, NJ, USA
| | - Renuka Kumar
- Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Nathan T Fried
- Department of Biology, Rutgers University, Camden, NJ, USA; Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA
| | - Tamara Hala
- Department of Biology, Rutgers University, Camden, NJ, USA
| | - Brian F Corbett
- Department of Biology, Rutgers University, Camden, NJ, USA; Center for Computational and Integrative Biology, Rutgers University, Camden, NJ, USA.
| |
Collapse
|
3
|
Isaac J, Murugan M. Interconnected neural circuits mediating social reward. Trends Neurosci 2024; 47:1041-1054. [PMID: 39532581 PMCID: PMC11633286 DOI: 10.1016/j.tins.2024.10.004] [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: 05/17/2024] [Revised: 08/26/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Across species, social behaviors are shaped and maintained through positive reinforcement of affiliative social interactions. As with nonsocial rewards, the reinforcing properties of social interactions have been shown to involve interplay between various brain regions and the mesolimbic reward system. In this review, we summarize findings from rodent research on the neural circuits that encode and mediate different components of social reward-seeking behavior. We explore methods to parse and study social reward-related behaviors using available behavioral paradigms. We also compare the neural mechanisms that support social versus nonsocial reward-seeking. Finally, we discuss how internal state and neuromodulatory systems affect reward-seeking behavior and the neural circuits that underlie social reward.
Collapse
Affiliation(s)
- Jennifer Isaac
- Neuroscience Graduate Program, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA
| | - Malavika Murugan
- Neuroscience Graduate Program, Emory University, Atlanta, GA 30322, USA; Department of Biology, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
4
|
Fricker BA, Murugan M, Seifert AW, Kelly AM. Cingulate to septal circuitry facilitates the preference to affiliate with large peer groups. Curr Biol 2024; 34:4452-4463.e4. [PMID: 39265570 PMCID: PMC11486304 DOI: 10.1016/j.cub.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Despite the prevalence of large-group living across the animal kingdom, no studies have examined the neural mechanisms that make group living possible. Spiny mice, Acomys, have evolved to live in large groups and exhibit a preference to affiliate with large over small groups. Here, we determine the neural circuitry that facilitates the drive to affiliate with large groups. We first identify an anterior cingulate cortex (ACC) to lateral septum (LS) circuit that is more responsive to large than small groups of novel same-sex peers. Using chemogenetics, we then demonstrate that this circuit is necessary for both male and female group investigation preferences but only males' preference to affiliate with larger peer groups. Furthermore, inhibition of the ACC-LS circuit specifically impairs social, but not nonsocial, affiliative grouping preferences. These findings reveal a key circuit for the regulation of mammalian peer group affiliation.
Collapse
Affiliation(s)
- Brandon A Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Malavika Murugan
- Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 211 Thomas Hunt Morgan Building, Lexington, KY 40506, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA.
| |
Collapse
|
5
|
Skandalakis GP, Neudorfer C, Payne CA, Bond E, Tavakkoli AD, Barrios-Martinez J, Trutti AC, Koutsarnakis C, Coenen VA, Komaitis S, Hadjipanayis CG, Stranjalis G, Yeh FC, Banihashemi L, Hong J, Lozano AM, Kogan M, Horn A, Evans LT, Kalyvas A. Establishing connectivity through microdissections of midbrain stimulation-related neural circuits. Brain 2024; 147:3083-3098. [PMID: 38808482 PMCID: PMC11370807 DOI: 10.1093/brain/awae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/15/2024] [Accepted: 04/21/2024] [Indexed: 05/30/2024] Open
Abstract
Comprehensive understanding of the neural circuits involving the ventral tegmental area is essential for elucidating the anatomofunctional mechanisms governing human behaviour, in addition to the therapeutic and adverse effects of deep brain stimulation for neuropsychiatric diseases. Although the ventral tegmental area has been targeted successfully with deep brain stimulation for different neuropsychiatric diseases, the axonal connectivity of the region is not fully understood. Here, using fibre microdissections in human cadaveric hemispheres, population-based high-definition fibre tractography and previously reported deep brain stimulation hotspots, we find that the ventral tegmental area participates in an intricate network involving the serotonergic pontine nuclei, basal ganglia, limbic system, basal forebrain and prefrontal cortex, which is implicated in the treatment of obsessive-compulsive disorder, major depressive disorder, Alzheimer's disease, cluster headaches and aggressive behaviours.
Collapse
Affiliation(s)
- Georgios P Skandalakis
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Clemens Neudorfer
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Caitlin A Payne
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Evalina Bond
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Armin D Tavakkoli
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | | | - Anne C Trutti
- Integrative Model-Based Cognitive Neuroscience Research Unit, University of Amsterdam, Amsterdam 15926, The Netherlands
| | - Christos Koutsarnakis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Volker A Coenen
- Department of Stereotactic and Functional Neurosurgery, Medical Center of the University of Freiburg, Freiburg 79106, Germany
- Medical Faculty of the University of Freiburg, Freiburg 79110, Germany
- Center for Deep Brain Stimulation, Medical Center of the University of Freiburg, Freiburg 79106, Germany
| | - Spyridon Komaitis
- Queens Medical Center, Nottingham University Hospitals NHS Foundation Trust, Nottingham NG7 2UH, UK
| | | | - George Stranjalis
- Department of Neurosurgery, National and Kapodistrian University of Athens Medical School, Evangelismos General Hospital, Athens 10676, Greece
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Layla Banihashemi
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jennifer Hong
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Andres M Lozano
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Michael Kogan
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM 87106, USA
| | - Andreas Horn
- Center for Brain Circuit Therapeutics Department of Neurology Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- MGH Neurosurgery & Center for Neurotechnology and Neurorecovery (CNTR) at MGH Neurology Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Movement Disorder and Neuromodulation Unit, Department of Neurology, Department of Neurology, Charité—Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Linton T Evans
- Section of Neurosurgery, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756, USA
| | - Aristotelis Kalyvas
- Division of Neurosurgery, University Health Network, University of Toronto, Toronto, ON M5T 1P5, Canada
| |
Collapse
|
6
|
Gershman SJ, Assad JA, Datta SR, Linderman SW, Sabatini BL, Uchida N, Wilbrecht L. Explaining dopamine through prediction errors and beyond. Nat Neurosci 2024; 27:1645-1655. [PMID: 39054370 DOI: 10.1038/s41593-024-01705-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/13/2024] [Indexed: 07/27/2024]
Abstract
The most influential account of phasic dopamine holds that it reports reward prediction errors (RPEs). The RPE-based interpretation of dopamine signaling is, in its original form, probably too simple and fails to explain all the properties of phasic dopamine observed in behaving animals. This Perspective helps to resolve some of the conflicting interpretations of dopamine that currently exist in the literature. We focus on the following three empirical challenges to the RPE theory of dopamine: why does dopamine (1) ramp up as animals approach rewards, (2) respond to sensory and motor features and (3) influence action selection? We argue that the prediction error concept, once it has been suitably modified and generalized based on an analysis of each computational problem, answers each challenge. Nonetheless, there are a number of additional empirical findings that appear to demand fundamentally different theoretical explanations beyond encoding RPE. Therefore, looking forward, we discuss the prospects for a unifying theory that respects the diversity of dopamine signaling and function as well as the complex circuitry that both underlies and responds to dopaminergic transmission.
Collapse
Affiliation(s)
- Samuel J Gershman
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA.
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA.
| | - John A Assad
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | | | - Scott W Linderman
- Department of Statistics and Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Bernardo L Sabatini
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Naoshige Uchida
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Linda Wilbrecht
- Department of Psychology and Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
7
|
Amada N, Sato S, Ishikawa D, Nakamura M, Suzuki M, Futamura T, Maeda K. Brexpiprazole: A new option in treating agitation in Alzheimer's dementia-Insights from transgenic mouse models. Neuropsychopharmacol Rep 2024; 44:557-568. [PMID: 38924384 PMCID: PMC11544443 DOI: 10.1002/npr2.12461] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
AIM Brexpiprazole is the first FDA-approved treatment for agitation associated with dementia due to Alzheimer's disease. Agitation in Alzheimer's dementia (AAD) occurs in high prevalence and is a great burden for patients and caregivers. Efficacy, safety, and tolerability of brexpiprazole were demonstrated in the AAD clinical trials. To demonstrate the agitation-ameliorating effect of brexpiprazole in animals, we evaluated brexpiprazole in two AAD mouse models. METHODS The resident-intruder test was conducted in 5- to 6-month-old Tg2576 mice, given vehicle or brexpiprazole (0.01 or 0.03 mg/kg) orally 1 h before the test. Locomotor activity was measured in 6-month-old APPSL-Tg mice given vehicle or brexpiprazole (0.01 or 0.03 mg/kg) orally the evening before the start of locomotor measurement for 3 days. RESULTS In the resident-intruder test, Tg2576 mice showed significantly higher attack number and shorter latency to first attack compared to non-Tg mice. In the Tg mice, brexpiprazole treatment (0.03 mg/kg) significantly delayed the latency to first attack and showed a trend toward a decrease in attack number. APPSL-Tg mice (≧6 months old) showed significantly higher locomotion during dark period Phase II (Zeitgeber time [ZT] 16-20) and Phase III (ZT20-24) compared to non-Tg mice, correlating with the clinical observations of late afternoon agitation in Alzheimer's disease. Brexpiprazole treatment (0.01 and 0.03 mg/kg) significantly decreased hyperlocomotion during the Phase III in APPSL-Tg mice. CONCLUSION The suppression of attack behavior and the reduction of nocturnal hyperlocomotion in these Tg mice may be indicative of the therapeutic effect of brexpiprazole on AAD, as demonstrated in the clinical trials.
Collapse
Affiliation(s)
- Naoki Amada
- Otsuka Pharmaceutical Co., Ltd.TokushimaJapan
| | | | | | | | | | | | | |
Collapse
|
8
|
Choi TY, Jeong S, Koo JW. Mesocorticolimbic circuit mechanisms of social dominance behavior. Exp Mol Med 2024; 56:1889-1899. [PMID: 39218974 PMCID: PMC11447232 DOI: 10.1038/s12276-024-01299-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/10/2024] [Accepted: 05/23/2024] [Indexed: 09/04/2024] Open
Abstract
Social animals, including rodents, primates, and humans, partake in competition for finite resources, thereby establishing social hierarchies wherein an individual's social standing influences diverse behaviors. Understanding the neurobiological underpinnings of social dominance is imperative, given its ramifications for health, survival, and reproduction. Social dominance behavior comprises several facets, including social recognition, social decision-making, and actions, indicating the concerted involvement of multiple brain regions in orchestrating this behavior. While extensive research has been dedicated to elucidating the neurobiology of social interaction, recent studies have increasingly delved into adverse social behaviors such as social competition and hierarchy. This review focuses on the latest advancements in comprehending the mechanisms of the mesocorticolimbic circuit governing social dominance, with a specific focus on rodent studies, elucidating the intricate dynamics of social hierarchies and their implications for individual well-being and adaptation.
Collapse
Affiliation(s)
- Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.
| | - Sejin Jeong
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
- Department of Life Sciences, Yeungnam University, Gyeongsan, Republic of Korea
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
9
|
Zhong CC, Xu Z, Gan J, Yu YM, Tang HM, Zhu Y, Yang JX, Ding HL, Cao JL. Acute Ongoing Nociception Delays Recovery of Consciousness from Sevoflurane Anesthesia via a Midbrain Circuit. J Neurosci 2024; 44:e0740242024. [PMID: 39019613 PMCID: PMC11340287 DOI: 10.1523/jneurosci.0740-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/07/2024] [Accepted: 07/10/2024] [Indexed: 07/19/2024] Open
Abstract
Although anesthesia provides favorable conditions for surgical procedures, recent studies have revealed that the brain remains active in processing noxious signals even during anesthesia. However, whether and how these responses affect the anesthesia effect remains unclear. The ventrolateral periaqueductal gray (vlPAG), a crucial hub for pain regulation, also plays an essential role in controlling general anesthesia. Hence, it was hypothesized that the vlPAG may be involved in the regulation of general anesthesia by noxious stimuli. Here, we found that acute noxious stimuli, including capsaicin-induced inflammatory pain, acetic acid-induced visceral pain, and incision-induced surgical pain, significantly delayed recovery from sevoflurane anesthesia in male mice, whereas this effect was absent in the spared nerve injury-induced chronic pain. Pretreatment with peripheral analgesics could prevent the delayed recovery induced by acute nociception. Furthermore, we found that acute noxious stimuli, induced by the injection of capsaicin under sevoflurane anesthesia, increased c-Fos expression and activity in the GABAergic neurons of the ventrolateral periaqueductal gray. Specific reactivation of capsaicin-activated vlPAGGABA neurons mimicked the effect of capsaicin and its chemogenetic inhibition prevented the delayed recovery from anesthesia induced by capsaicin. Finally, we revealed that the vlPAGGABA neurons regulated the recovery from anesthesia through the inhibition of ventral tegmental area dopaminergic neuronal activity, thus decreasing dopamine (DA) release and activation of DA D1-like receptors in the brain. These findings reveal a novel, cell- and circuit-based mechanism for regulating anesthesia recovery by nociception, and it is important to provide new insights for guiding the management of the anesthesia recovery period.
Collapse
Affiliation(s)
- Chao-Chao Zhong
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Zheng Xu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun Gan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yu-Mei Yu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Hui-Mei Tang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Yangzi Zhu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun-Xia Yang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Hai-Lei Ding
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
| | - Jun-Li Cao
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou 221004, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou 221004, China
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou 221004, China
- Department of Anesthesiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China
| |
Collapse
|
10
|
Zhang S, Zhang X, Li H, Wang D, Wang S, Wang Y, Zhao G, Dong H, Li J. Ventral Tegmental Area Glutamatergic Neurons Facilitated Emergence From Isoflurane Anesthesia Involves Excitation of Lateral Septum GABA-ergic Neurons in Mice. Anesth Analg 2024; 139:397-410. [PMID: 38048607 DOI: 10.1213/ane.0000000000006739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
BACKGROUND Ventral tegmental area (VTA) glutamatergic neurons promote wakefulness in the sleep-wake cycle; however, their roles and neural circuit mechanisms during isoflurane (ISO) anesthesia remain unclear. METHODS Fiber photometry and in vivo electrophysiology were used to observe the changes in neuronal or terminal activity during ISO anesthesia and arousal processes. Optogenetic and anesthesia behaviors were used to investigate the effects of VTA glutamatergic neurons and their projections to the lateral septum (LS) during ISO anesthesia and arousal. Anterograde and retrograde tracings were performed to identify the connections between VTA glutamatergic neurons and the LS. RESULTS Population activity and firing rates of VTA glutamatergic neurons decreased during ISO anesthesia (ISO: 95% confidence interval [CI], 0.83-2.06 Spikes.s -1 vs wake: 95% CI, 3.53-7.83 Spikes.s -1 ; P =.0001; n = 34 from 4 mice). Optogenetic activation of VTA glutamatergic neurons reduced the burst-suppression ratio in electroencephalography (laser: 95% CI, 13.09%-28.76% vs pre: 95% CI, 52.85%-71.59%; P =.0009; n = 6) and facilitated emergence (ChR2: 95% CI, 343.3-388.0 seconds vs mCherry: 95% CI, 447.6-509.8 seconds; P < .0001; n = 11/12) from ISO anesthesia. VTA glutamatergic neurons monosynaptically innervated LS γ-aminobutyric acid (GABA)-ergic neurons. The activity of VTA glutamatergic terminals in the LS decreased during ISO anesthesia, and optogenetic activation of the VTA glutamatergic terminals in the LS facilitated emergence from ISO anesthesia. Furthermore, optogenetic activation of VTA glutamatergic terminals increased the firing rates of LS γ-aminobutyric acid-ergic (GABAergic) neurons (laser: 95% CI, 0.85-4.03 Spikes.s -1 vs pre: 95% CI, 0.24-0.78 Spikes.s -1 ; P =.008; n = 23 from 4 mice) during ISO anesthesia. CONCLUSIONS VTA glutamatergic neurons facilitated emergence from ISO anesthesia involving excitation of LS GABAergic neurons.
Collapse
Affiliation(s)
- Simin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinxin Zhang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huiming Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dan Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Sa Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuhao Wang
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Guangchao Zhao
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hailong Dong
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiannan Li
- From the Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
11
|
Fang YT, Kuo HC, Chen CY, Chou SJ, Lu CW, Hung CM. Brain Gene Regulatory Networks Coordinate Nest Construction in Birds. Mol Biol Evol 2024; 41:msae125. [PMID: 38916488 PMCID: PMC11223658 DOI: 10.1093/molbev/msae125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/18/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024] Open
Abstract
Nest building is a vital behavior exhibited during breeding in birds, and is possibly induced by environmental and social cues. Although such behavioral plasticity has been hypothesized to be controlled by adult neuronal plasticity, empirical evidence, especially at the neurogenomic level, remains limited. Here, we aim to uncover the gene regulatory networks that govern avian nest construction and examine whether they are associated with circuit rewiring. We designed an experiment to dissect this complex behavior into components in response to pair bonding and nest material acquisition by manipulating the presence of mates and nest materials in 30 pairs of zebra finches. Whole-transcriptome analysis of 300 samples from five brain regions linked to avian nesting behaviors revealed nesting-associated gene expression enriched with neural rewiring functions, including neurogenesis and neuron projection. The enriched expression was observed in the motor/sensorimotor and social behavior networks of female finches, and in the dopaminergic reward system of males. Female birds exhibited predominant neurotranscriptomic changes to initiate the nesting stage, while males showed major changes after entering this stage, underscoring sex-specific roles in nesting behavior. Notably, major neurotranscriptomic changes occurred during pair bonding, with minor changes during nest material acquisition, emphasizing social interactions in nest construction. We also revealed gene expression associated with reproductive behaviors and tactile sensing for nesting behavior. This study presents novel neurogenomic evidence supporting the hypothesis of adult neural plasticity underlying avian nest-construction behavior. By uncovering the genetic toolkits involved, we offer novel insights into the evolution of animals' innate ability to construct nests.
Collapse
Affiliation(s)
- Yi-Ting Fang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Hao-Chih Kuo
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Yu Chen
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Shen-Ju Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-Wei Lu
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| | - Chih-Ming Hung
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
12
|
Baek S, Jang J, Jung HJ, Lee H, Choe Y. Advanced Immunolabeling Method for Optical Volumetric Imaging Reveals Dystrophic Neurites of Dopaminergic Neurons in Alzheimer's Disease Mouse Brain. Mol Neurobiol 2024; 61:3976-3999. [PMID: 38049707 PMCID: PMC11236860 DOI: 10.1007/s12035-023-03823-9] [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: 05/25/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023]
Abstract
Optical brain clearing combined with immunolabeling is valuable for analyzing molecular tissue structures, including complex synaptic connectivity. However, the presence of aberrant lipid deposition due to aging and brain disorders poses a challenge for achieving antibody penetration throughout the entire brain volume. Herein, we present an efficient brain-wide immunolabeling method, the immuno-active clearing technique (iACT). The treatment of brain tissues with a zwitterionic detergent, specifically SB3-12, significantly enhanced tissue permeability by effectively mitigating lipid barriers. Notably, Quadrol treatment further refines the methodology by effectively eliminating residual detergents from cleared brain tissues, subsequently amplifying volumetric fluorescence signals. Employing iACT, we uncover disrupted axonal projections within the mesolimbic dopaminergic (DA) circuits in 5xFAD mice. Subsequent characterization of DA neural circuits in 5xFAD mice revealed proximal axonal swelling and misrouting of distal axonal compartments in proximity to amyloid-beta plaques. Importantly, these structural anomalies in DA axons correlate with a marked reduction in DA release within the nucleus accumbens. Collectively, our findings highlight the efficacy of optical volumetric imaging with iACT in resolving intricate structural alterations in deep brain neural circuits. Furthermore, we unveil the compromised integrity of DA pathways, contributing to the underlying neuropathology of Alzheimer's disease. The iACT technique thus holds significant promise as a valuable asset for advancing our understanding of complex neurodegenerative disorders and may pave the way for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Soonbong Baek
- Developmental Disorders & Rare Diseases Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu, 41062, Republic of Korea
| | - Jaemyung Jang
- Developmental Disorders & Rare Diseases Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu, 41062, Republic of Korea
| | - Hyun Jin Jung
- Developmental Disorders & Rare Diseases Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu, 41062, Republic of Korea
| | - Hyeyoung Lee
- Division of Applied Bioengineering, Dong-eui University, Busanjin-gu, Busan, 47340, Republic of Korea
| | - Youngshik Choe
- Developmental Disorders & Rare Diseases Research Group, Korea Brain Research Institute, 61 Cheomdan-ro, Daegu, 41062, Republic of Korea.
| |
Collapse
|
13
|
Peng S, Yang X, Meng S, Liu F, Lv Y, Yang H, Kong Y, Xie W, Li M. Dual circuits originating from the ventral hippocampus independently facilitate affective empathy. Cell Rep 2024; 43:114277. [PMID: 38805397 DOI: 10.1016/j.celrep.2024.114277] [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: 07/26/2023] [Revised: 03/24/2024] [Accepted: 05/09/2024] [Indexed: 05/30/2024] Open
Abstract
Affective empathy enables social mammals to learn and transfer emotion to conspecifics, but an understanding of the neural circuitry and genetics underlying affective empathy is still very limited. Here, using the naive observational fear between cagemates as a paradigm similar to human affective empathy and chemo/optogenetic neuroactivity manipulation in mouse brain, we investigate the roles of multiple brain regions in mouse affective empathy. Remarkably, two neural circuits originating from the ventral hippocampus, previously unknown to function in empathy, are revealed to regulate naive observational fear. One is from ventral hippocampal pyramidal neurons to lateral septum GABAergic neurons, and the other is from ventral hippocampus pyramidal neurons to nucleus accumbens dopamine-receptor-expressing neurons. Furthermore, we identify the naive observational-fear-encoding neurons in the ventral hippocampus. Our findings highlight the potentially diverse regulatory pathways of empathy in social animals, shedding light on the mechanisms underlying empathy circuity and its disorders.
Collapse
Affiliation(s)
- Siqi Peng
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Xiuqi Yang
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Sibie Meng
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Fuyuan Liu
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China
| | - Yaochen Lv
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Huiquan Yang
- School of Medicine, Southeast University, Nanjing 210096, China
| | - Youyong Kong
- Jiangsu Provincial Joint International Research Laboratory of Medical Information Processing, School of Computer Science and Engineering, Southeast University, Nanjing 210096, China
| | - Wei Xie
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China; Jiangsu Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China
| | - Moyi Li
- School of Life Science and Technology, The Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China; Jiangsu Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China.
| |
Collapse
|
14
|
Zhang L, Sun Y, Wang J, Zhang M, Wang Q, Xie B, Yu F, Wen D, Ma C. Dopaminergic dominance in the ventral medial hypothalamus: A pivotal regulator for methamphetamine-induced pathological aggression. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110971. [PMID: 38365104 DOI: 10.1016/j.pnpbp.2024.110971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/05/2024] [Accepted: 02/13/2024] [Indexed: 02/18/2024]
Abstract
Methamphetamine (METH) abuse is associated with a spectrum of behavioral consequences, among which heightened aggression presents a significant challenge. However, the causal role of METH's impact in aggression and its target circuit mechanisms remains largely unknown. We established an acute METH exposure-aggression mouse model to investigate the role of ventral tegmental area (VTA) dopaminergic neurons and ventral medial hypothalamus VMH glutamatergic neuron. Our findings revealed that METH-induced VTA dopamine excitability activates the ventromedial hypothalamus (VMH) glutamatergic neurons, contributing to pathological aggression. Notably, we uncovered a dopaminergic transmission within the VTA-VMH circuit that exclusively functioned under METH influence. This dopaminergic pathway emerged as a potential key player in enabling dopamine-related pathological aggression, with heightened dopaminergic excitability implicated in various psychiatric symptoms. Also, the modulatory function of this pathway opens new possibilities for targeted therapeutic strategies for intervention to improve treatment in METH abuse and may have broader implications for addressing pathological aggression syndromes.
Collapse
Affiliation(s)
- Ludi Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China; Hebei Medical University Postdoctoral Research Station, 050017, Shijiazhuang, Hebei, PR China
| | - Yufei Sun
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Jian Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Minglong Zhang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Qingwu Wang
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China
| | - Di Wen
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Identification Center of Forensic Medicine, Hebei Medical University, 050017 Shijiazhuang, Hebei, PR China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017 Shijiazhuang, Hebei, PR China.
| |
Collapse
|
15
|
Puska G, Szendi V, Dobolyi A. Lateral septum as a possible regulatory center of maternal behaviors. Neurosci Biobehav Rev 2024; 161:105683. [PMID: 38649125 DOI: 10.1016/j.neubiorev.2024.105683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
The lateral septum (LS) is involved in controlling anxiety, aggression, feeding, and other motivated behaviors. Lesion studies have also implicated the LS in various forms of caring behaviors. Recently, novel experimental tools have provided a more detailed insight into the function of the LS, including the specific role of distinct cell types and their neuronal connections in behavioral regulations, in which the LS participates. This article discusses the regulation of different types of maternal behavioral alterations using the distributions of established maternal hormones such as prolactin, estrogens, and the neuropeptide oxytocin. It also considers the distribution of neurons activated in mothers in response to pups and other maternal activities, as well as gene expressional alterations in the maternal LS. Finally, this paper proposes further research directions to keep up with the rapidly developing knowledge on maternal behavioral control in other maternal brain regions.
Collapse
Affiliation(s)
- Gina Puska
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Department of Zoology, University of Veterinary Medicine Budapest, Budapest, Hungary
| | - Vivien Szendi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Arpád Dobolyi
- Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary; Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
16
|
Illera JC, Jimenez-Blanco F, Centenera L, Gil-Cabrera F, Crespo B, Lopez PR, Silvan G, Caceres S. Addressing Combative Behaviour in Spanish Bulls by Measuring Hormonal Indicators. Vet Sci 2024; 11:182. [PMID: 38668449 PMCID: PMC11053816 DOI: 10.3390/vetsci11040182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/02/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
The fighting bull is characterised by its natural aggressiveness, but the physiological mechanisms that underlie its aggressive behaviour are poorly studied. This study determines the hormonal component of aggressiveness in fighting bulls by analysing their behaviour during a fight and correlating it to their serotonin, dopamine and testosterone levels. We also determine whether aggressive behaviour can be estimated in calves. Using 195 animals, samples were obtained when the animals were calves and after 5 years. Aggressiveness scores were obtained by an observational method during bullfights, and serotonin, dopamine and testosterone levels were determined in all animals using validated enzyme immunoassay kits. The results revealed a strong correlation of serotonin and dopamine levels with aggressiveness scores in bulls during fights, but no correlation was found with respect to testosterone. These correlations led to established cut-off point and linear regression curves to obtain expected aggressiveness scores for calves at shoeing. There were no significant differences between the expected scores obtained in calves and the observed scores in bulls. Therefore, this study demonstrates that hormone determination in calves may be a great indicator of combativeness in bulls and can reliably be used in the selection of fighting bulls.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gema Silvan
- Department Animal Physiology, Veterinary Medicine School, Complutense University of Madrid (UCM), 28040 Madrid, Spain; (J.C.I.); (F.J.-B.); (L.C.); (F.G.-C.); (B.C.); (P.R.L.); (S.C.)
| | | |
Collapse
|
17
|
Huang B, Liang S, Li X, Xie Z, Yang R, Sun B, Xue J, Li B, Wang S, Shi H, Shi Y. Postweaning intermittent sleep deprivation enhances defensive attack in adult female mice via the microbiota-gut-brain axis. Prog Neuropsychopharmacol Biol Psychiatry 2024; 130:110915. [PMID: 38104921 DOI: 10.1016/j.pnpbp.2023.110915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Sleep is one of the most important physiological activities in life and promotes the growth and development of an individual. In modern society, sleep deprivation (SD), especially among adolescents, has become a common phenomenon. However, long-term SD severely affected adolescents' neurodevelopment leading to abnormal behavioral phenotypes. Clinical studies indicated that sleep problems caused increased aggressive behavior in adolescents. Aggressive behavior was subordinate to social behaviors, in which defensive attack was often the last line for survival. Meanwhile, increasing studies shown that gut microbiota regulated social behaviors by affecting specific brain regions via the gut-brain axis. However, whether postweaning intermittent SD is related to defensive attack in adulthood, and if so, whether it is mediated by the microbiota-gut-brain axis are still elusive. Combined with microbial sequencing and hippocampal metabolomics, the present study mainly investigated the long-term effects of postweaning intermittent SD on defensive attack in adult mice. Our study demonstrated that postweaning intermittent SD enhanced defensive attack and impaired long-term memory formation in adult female mice. Moreover, microbial sequencing and LC-MS analysis showed that postweaning intermittent SD altered the gut microbial composition and the hippocampal metabolic profile in female mice, respectively. Our attention has been drawn to the neuroactive ligand-receptor interaction pathway and related metabolites. In conclusion, our findings provide a new perspective on the relationship of early-life SD and defensive attack in adulthood, and also highlight the importance of sleep in early-life, especially in females.
Collapse
Affiliation(s)
- Boya Huang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China; Graduate School, Tianjin Medical University, Tianjin 300070, China
| | - Shihao Liang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Xinrui Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Ziyu Xie
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui Yang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Binhuang Sun
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Jiping Xue
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Bingyu Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China
| | - Sheng Wang
- Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China; Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang 050017, China; Nursing School, Hebei Medical University, Shijiazhuang 050031, China.
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang 050017, China; Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medicinal University, Shijiazhuang 050017, China.
| |
Collapse
|
18
|
Veyrunes F, Perez J, Heitzmann LD, Saunders PA, Givalois L. Hormone profiles of the African pygmy mouse Mus minutoides, a species with XY female sex reversal. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:130-137. [PMID: 38059664 DOI: 10.1002/jez.2767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023]
Abstract
In mammals, most sex differences in phenotype are controlled by gonadal hormones, but recent work on transgenic mice has shown that sex chromosomes can have a direct influence on sex-specific behaviors. In this study, we take advantage of the naturally occurring sex reversal in a mouse species, Mus minutoides, to investigate for the first time the relationship between sex chromosomes, hormones, and behaviors in a wild species. In this model, a feminizing variant of the X chromosome, named X*, produces three types of females with different sex chromosome complements (XX, XX*, and X*Y), associated with alternative behavioral phenotypes, while all males are XY. We thus compared the levels of three major circulating steroid hormones (testosterone, corticosterone, and estradiol) in the four sex genotypes to disentangle the influence of sex chromosomes and sex hormones on behavior. First, we did not find any difference in testosterone levels in the three female genotypes, although X*Y females are notoriously more aggressive. Second, in agreement with their lower anxiety-related behaviors, X*Y females and XY males display lower baseline corticosterone concentration than XX and XX* females. Instead of a direct hormonal influence, this result rather suggests that sex chromosomes may have an impact on the baseline corticosterone level, which in turn may influence behaviors. Third, estradiol concentrations do not explain the enhanced reproductive performance and maternal care behavior of the X*Y females compared to the XX and XX* females. Overall, this study highlights that most of the behaviors varying along with sex chromosome complement of this species are more likely driven by genetic factors rather than steroid hormone concentrations.
Collapse
Affiliation(s)
- Frederic Veyrunes
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Julie Perez
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Louise D Heitzmann
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Paul A Saunders
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Laurent Givalois
- MMDN, Molecular Mechanisms in Neurodegenerative Dementia Laboratory, Université Montpellier, EPHE-PSL, INSERM U1198, Montpellier, France
- Department of Psychiatry and Neurosciences, CR-CHUQ, Faculty of Medicine, Laval University, Québec City, Canada
- CNRS, Paris, France
| |
Collapse
|
19
|
Rodriguez LA, Tran MN, Garcia-Flores R, Oh S, Phillips RA, Pattie EA, Divecha HR, Kim SH, Shin JH, Lee YK, Montoya C, Jaffe AE, Collado-Torres L, Page SC, Martinowich K. TrkB-dependent regulation of molecular signaling across septal cell types. Transl Psychiatry 2024; 14:52. [PMID: 38263132 PMCID: PMC10805920 DOI: 10.1038/s41398-024-02758-6] [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: 12/20/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
The lateral septum (LS), a GABAergic structure located in the basal forebrain, is implicated in social behavior, learning, and memory. We previously demonstrated that expression of tropomyosin kinase receptor B (TrkB) in LS neurons is required for social novelty recognition. To better understand molecular mechanisms by which TrkB signaling controls behavior, we locally knocked down TrkB in LS and used bulk RNA-sequencing to identify changes in gene expression downstream of TrkB. TrkB knockdown induces upregulation of genes associated with inflammation and immune responses, and downregulation of genes associated with synaptic signaling and plasticity. Next, we generated one of the first atlases of molecular profiles for LS cell types using single nucleus RNA-sequencing (snRNA-seq). We identified markers for the septum broadly, and the LS specifically, as well as for all neuronal cell types. We then investigated whether the differentially expressed genes (DEGs) induced by TrkB knockdown map to specific LS cell types. Enrichment testing identified that downregulated DEGs are broadly expressed across neuronal clusters. Enrichment analyses of these DEGs demonstrated that downregulated genes are uniquely expressed in the LS, and associated with either synaptic plasticity or neurodevelopmental disorders. Upregulated genes are enriched in LS microglia, associated with immune response and inflammation, and linked to both neurodegenerative disease and neuropsychiatric disorders. In addition, many of these genes are implicated in regulating social behaviors. In summary, the findings implicate TrkB signaling in the LS as a critical regulator of gene networks associated with psychiatric disorders that display social deficits, including schizophrenia and autism, and with neurodegenerative diseases, including Alzheimer's.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Renee Garcia-Flores
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Seyun Oh
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Robert A Phillips
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Yong Kyu Lee
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Carly Montoya
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
20
|
Yadav RSP, Ansari F, Bera N, Kent C, Agrawal P. Lessons from lonely flies: Molecular and neuronal mechanisms underlying social isolation. Neurosci Biobehav Rev 2024; 156:105504. [PMID: 38061597 DOI: 10.1016/j.neubiorev.2023.105504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/26/2023]
Abstract
Animals respond to changes in the environment which affect their internal state by adapting their behaviors. Social isolation is a form of passive environmental stressor that alters behaviors across animal kingdom, including humans, rodents, and fruit flies. Social isolation is known to increase violence, disrupt sleep and increase depression leading to poor mental and physical health. Recent evidences from several model organisms suggest that social isolation leads to remodeling of the transcriptional and epigenetic landscape which alters behavioral outcomes. In this review, we explore how manipulating social experience of fruit fly Drosophila melanogaster can shed light on molecular and neuronal mechanisms underlying isolation driven behaviors. We discuss the recent advances made using the powerful genetic toolkit and behavioral assays in Drosophila to uncover role of neuromodulators, sensory modalities, pheromones, neuronal circuits and molecular mechanisms in mediating social isolation. The insights gained from these studies could be crucial for developing effective therapeutic interventions in future.
Collapse
Affiliation(s)
- R Sai Prathap Yadav
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Faizah Ansari
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Neha Bera
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India
| | - Clement Kent
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Pavan Agrawal
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal Academy of Higher Education, Karnataka 576104, India.
| |
Collapse
|
21
|
Song BL, Zhou J, Jiang Y, Li LF, Liu YJ. Dopamine D2 receptor within the intermediate region of the lateral septum modulate social hierarchy in male mice. Neuropharmacology 2023; 241:109735. [PMID: 37788799 DOI: 10.1016/j.neuropharm.2023.109735] [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: 07/05/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/05/2023]
Abstract
The dopamine (DA) system has long been involved in social hierarchies; however, the specific mechanisms have not been elucidated. The lateral septum (LS) is a limbic brain structure that regulates various emotional, motivational, and social behaviors. DA receptors are abundantly expressed in the LS, modulating its functions. In this study, we evaluated the functions of DA receptors within different subregions of the LS in social dominance using a confrontation tube test in male mice. The results showed that mice living in social groups formed linear dominance hierarchies after a few days of cohousing, and the subordinates showed increased anxiety. Fos expressions was elevated in the entire LS after a confrontation tube test in the subordinates. However, DA neurons were more activated in the dominates within the ventral tegmental area and the dorsal raphe nucleus. Quantitative real-time polymerase chain reaction results showed that D2 receptor (D2R) within the intermediate region of the LS (LSi) were elevated in the subordinate. In the following pharmacological studies, we found simultaneous D2R activation in the dominants and D2R inhibition in the subordinates switched the original dominant-subordinate relationship. The aforementioned results suggested that D2R within the LSi plays an important role in social dominance in male mice. These findings improve our understanding of the neural mechanisms underlying the social hierarchy, which is closely related to our social life and happiness.
Collapse
Affiliation(s)
- Bai-Lin Song
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Jie Zhou
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Yi Jiang
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China
| | - Lai-Fu Li
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China.
| | - Ying-Juan Liu
- Research Center of Henan Provincial Agricultural Biomass Resource Engineering and Technology, College of Life Science and Agriculture, Nanyang Normal University, Nanyang, 473061, China.
| |
Collapse
|
22
|
Stagkourakis S, Spigolon G, Marks M, Feyder M, Kim J, Perona P, Pachitariu M, Anderson DJ. Anatomically distributed neural representations of instincts in the hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568163. [PMID: 38045312 PMCID: PMC10690204 DOI: 10.1101/2023.11.21.568163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Artificial activation of anatomically localized, genetically defined hypothalamic neuron populations is known to trigger distinct innate behaviors, suggesting a hypothalamic nucleus-centered organization of behavior control. To assess whether the encoding of behavior is similarly anatomically confined, we performed simultaneous neuron recordings across twenty hypothalamic regions in freely moving animals. Here we show that distinct but anatomically distributed neuron ensembles encode the social and fear behavior classes, primarily through mixed selectivity. While behavior class-encoding ensembles were spatially distributed, individual ensembles exhibited strong localization bias. Encoding models identified that behavior actions, but not motion-related variables, explained a large fraction of hypothalamic neuron activity variance. These results identify unexpected complexity in the hypothalamic encoding of instincts and provide a foundation for understanding the role of distributed neural representations in the expression of behaviors driven by hardwired circuits.
Collapse
Affiliation(s)
- Stefanos Stagkourakis
- Division of Biology and Biological Engineering 156-29, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California 91125, USA
| | - Giada Spigolon
- Biological Imaging Facility, California Institute of Technology, Pasadena, California 91125, USA
| | - Markus Marks
- Division of Engineering and Applied Science, California Institute of Technology, Pasadena, California 91125, USA
| | - Michael Feyder
- Division of Psychiatry and Behavioral Sciences, Stanford University, Palo Alto, California 94305, USA
| | - Joseph Kim
- Division of Biology and Biological Engineering 156-29, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California 91125, USA
| | - Pietro Perona
- Division of Engineering and Applied Science, California Institute of Technology, Pasadena, California 91125, USA
| | - Marius Pachitariu
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - David J. Anderson
- Division of Biology and Biological Engineering 156-29, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, California 91125, USA
- Howard Hughes Medical Institute, California Institute of Technology, 1200 East California Blvd, Pasadena, California 91125, USA
| |
Collapse
|
23
|
Blaess S, Krabbe S. Cell type specificity for circuit output in the midbrain dopaminergic system. Curr Opin Neurobiol 2023; 83:102811. [PMID: 37972537 DOI: 10.1016/j.conb.2023.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Midbrain dopaminergic neurons are a relatively small group of neurons in the mammalian brain controlling a wide range of behaviors. In recent years, increasingly sophisticated tracing, imaging, transcriptomic, and machine learning approaches have provided substantial insights into the anatomical, molecular, and functional heterogeneity of dopaminergic neurons. Despite this wealth of new knowledge, it remains unclear whether and how the diverse features defining dopaminergic subclasses converge to delineate functional ensembles within the dopaminergic system. Here, we review recent studies investigating various aspects of dopaminergic heterogeneity and discuss how development, behavior, and disease influence subtype characteristics. We then outline what further approaches could be pursued to gain a more inclusive picture of dopaminergic diversity, which could be crucial to understanding the functional architecture of this system.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Sabine Krabbe
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
24
|
Dromard Y, Borie AM, Chakraborty P, Muscatelli F, Guillon G, Desarménien MG, Jeanneteau F. Disengagement of somatostatin neurons from lateral septum circuitry by oxytocin and vasopressin restores social-fear extinction and suppresses aggression outbursts in Prader-Willi syndrome model. Biol Psychiatry 2023; 95:S0006-3223(23)01661-X. [PMID: 39491230 PMCID: PMC11216544 DOI: 10.1016/j.biopsych.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 07/03/2024]
Abstract
BACKGROUND Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. METHODS Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. RESULTS OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. CONCLUSIONS SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.
Collapse
Affiliation(s)
- Yann Dromard
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Amélie M Borie
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Prabahan Chakraborty
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Françoise Muscatelli
- Institut de Neurobiologie de la Méditerranée, INSERM, University of Aix-Marseille, Marseille, France
| | - Gilles Guillon
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Michel G Desarménien
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France
| | - Freddy Jeanneteau
- Institut de Génomique Fonctionnelle, University of Montpellier, INSERM, CNRS, France.
| |
Collapse
|
25
|
Fricker BA, Ho D, Seifert AW, Kelly AM. Biased brain and behavioral responses towards kin in males of a communally breeding species. Sci Rep 2023; 13:17040. [PMID: 37813903 PMCID: PMC10562393 DOI: 10.1038/s41598-023-44257-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
In complex social environments, individuals may interact with not only novel and familiar conspecifics but also kin and non-kin. The ability to distinguish between conspecific identities is crucial for most animals, yet how the brain processes conspecific type and how animals may alter behavior accordingly is not well known. We examined whether the communally breeding spiny mouse (Acomys cahirinus) responds differently to conspecifics that vary in novelty and kinship. In a group interaction test, we found that males can distinguish novel kin from novel non-kin, and preferentially spend time with novel kin over familiar kin and novel non-kin. To determine whether kinship and novelty status are differentially represented in the brain, we conducted immediate early gene tests, which revealed the dorsal, but not ventral, lateral septum differentially processes kinship. Neither region differentially processes social novelty. Further, males did not exhibit differences in prosocial behavior toward novel and familiar conspecifics but exhibited more prosocial behavior with novel kin than novel non-kin. These results suggest that communally breeding species may have evolved specialized neural circuitry to facilitate a bias to be more affiliative with kin, regardless of whether they are novel or familiar, potentially to promote prosocial behaviors, thereby facilitating group cohesion.
Collapse
Affiliation(s)
- Brandon A Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - Deborah Ho
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 101 Morgan Building, Lexington, KY, 40506, USA
| | - Aubrey M Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA, 30322, USA.
| |
Collapse
|
26
|
Cui X, Tong Q, Xu H, Xie C, Xiao L. A putative loop connection between VTA dopamine neurons and nucleus accumbens encodes positive valence to compensate for hunger. Prog Neurobiol 2023; 229:102503. [PMID: 37451329 DOI: 10.1016/j.pneurobio.2023.102503] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Dopamine (DA) signal play pivotal roles in regulating motivated behaviors, including feeding behavior, but the role of midbrain DA neurons in modulating food intake and neural circuitry mechanisms remain largely unknown. Here, we found that activating but not inhibiting ventral tegmental area (VTA) DA neurons reduces mouse food intake. Furthermore, DA neurons in ventral VTA, especially neurons projecting to the medial nucleus accumbens (NAc), are activated by refeeding in the 24 h fasted mice. Combing neural circuitry tracing, optogenetic, chemogenetic, and pharmacological manipulations, we established that the VTA→medial NAc→VTA loop circuit is critical for the VTA DA neurons activation-induced food intake reduction. Moreover, activating either VTA DA neurons or dopaminergic axons in medial NAc elevates positive valence, which will compensate for the hungry-induced food intake. Thus, our study identifies a subset of positive valence-encoded VTA DA neurons forming possible loop connections with medial NAc that are anorexigenic.
Collapse
Affiliation(s)
- Xiao Cui
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qiuping Tong
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Hao Xu
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Chuantong Xie
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Lei Xiao
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
27
|
Xu C, Wang F, Huang Q, Lyu D, Wu C, Cao T, Zhao J, Wang M, Zhou N, Yang W, Chen Y, Wei Z, Xie B, Hong W. Association between overt aggression and anhedonia in patients with major depressive disorder during the acute phase. J Psychiatr Res 2023; 165:41-47. [PMID: 37459777 DOI: 10.1016/j.jpsychires.2023.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 07/04/2023] [Accepted: 07/10/2023] [Indexed: 09/03/2023]
Abstract
OBJECTIVE To explore the factors influencing anhedonia at baseline and use them as confounding factors. To further investigate the correlation between overt aggression and anhedonia during the acute phase of major depressive disorder. METHODS In this eight-week prospective study, 384 major depressive disorder patients were recruited from the outpatient section of Shanghai Mental Health Center from May 1, 2017, to October 30, 2018. Standard treatments were performed with escitalopram or venlafaxine for participants. Depressive symptoms, overt aggression, and anhedonia were assessed using the 17-item Hamilton Rating Scale for Depression, Modified Overt Aggression Scale, and Snaith-Hamilton Pleasure Scale at baseline, and in the 4th and 8th weeks. RESULTS Obsessive-compulsive symptoms and the duration of untreated psychosis were positively associated with aggression (P < 0.05). Patients with aggressive behaviour had worse cognitive impairment and severe anhedonia of pleasurable sensory experiences (P < 0.05). For anhedonia, being female (tau_b = -0.23, P = 0.012) was a protective factor, while number of recurrent, melancholic features, current obsessions, previous combination drug therapies, depressive symptoms, and aggressive behaviour were risk factors (P < 0.05). Social anhedonia related to interests/pastimes, and pleasurable sensory experiences were more severe in major depressive disorder patients with aggressive behaviour in the acute phase (P < 0.05). CONCLUSIONS Anhedonia persisted in major depressive disorder patients with aggressive behaviour after standardized treatment during the acute phase. Being female protected the pleasures from social interaction and sensory experience. However, the number of depressive episodes, melancholic features, current obsessive symptoms, previous combination drug therapies, depressive symptoms, and aggressive behaviour was positively associated with anhedonia.
Collapse
Affiliation(s)
- Chuchen Xu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Fan Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China. beauty--
| | - Qinte Huang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Dongbin Lyu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Chenglin Wu
- Shanghai Pudong New Area Mental Health Center, School of Medicine, Tongji University, Shanghai, 200124, China.
| | - Tongdan Cao
- Shanghai Mental Health Center of Huangpu District, Shanghai, 200011, China.
| | - Jie Zhao
- Shanghai Mental Health Center of Huangpu District, Shanghai, 200011, China.
| | - Meiti Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Ni Zhou
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Weichieh Yang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Yiming Chen
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Zheyi Wei
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Bin Xie
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 20030, China.
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; Shanghai Key Laboratory of Psychotic Disorders, Shanghai, 20030, China.
| |
Collapse
|
28
|
Suri D, Zanni G, Mahadevia D, Chuhma N, Saha R, Spivack S, Pini N, Stevens GS, Ziolkowski-Blake A, Simpson EH, Balsam P, Rayport S, Ansorge MS. Dopamine transporter blockade during adolescence increases adult dopamine function, impulsivity, and aggression. Mol Psychiatry 2023; 28:3512-3523. [PMID: 37532798 PMCID: PMC10618097 DOI: 10.1038/s41380-023-02194-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023]
Abstract
Sensitive developmental periods shape neural circuits and enable adaptation. However, they also engender vulnerability to factors that can perturb developmental trajectories. An understanding of sensitive period phenomena and mechanisms separate from sensory system development is still lacking, yet critical to understanding disease etiology and risk. The dopamine system is pivotal in controlling and shaping adolescent behaviors, and it undergoes heightened plasticity during that time, such that interference with dopamine signaling can have long-lasting behavioral consequences. Here we sought to gain mechanistic insight into this dopamine-sensitive period and its impact on behavior. In mice, dopamine transporter (DAT) blockade from postnatal (P) day 22 to 41 increases aggression and sensitivity to amphetamine (AMPH) behavioral stimulation in adulthood. Here, we refined this sensitive window to P32-41 and identified increased firing of dopaminergic neurons in vitro and in vivo as a neural correlate to altered adult behavior. Aggression can result from enhanced impulsivity and cognitive dysfunction, and dopamine regulates working memory and motivated behavior. Hence, we assessed these behavioral domains and found that P32-41 DAT blockade increases impulsivity but has no effect on cognition, working memory, or motivation in adulthood. Lastly, using optogenetics to drive dopamine neurons, we find that increased VTA but not SNc dopaminergic activity mimics the increase in impulsive behavior in the Go/NoGo task observed after adolescent DAT blockade. Together our data provide insight into the developmental origins of aggression and impulsivity that may ultimately improve diagnosis, prevention, and treatment strategies for related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Deepika Suri
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Giulia Zanni
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Darshini Mahadevia
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Nao Chuhma
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Rinki Saha
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Stephen Spivack
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Nicolò Pini
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Gregory S Stevens
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Annette Ziolkowski-Blake
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Eleanor H Simpson
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Peter Balsam
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA
- Department of Neuroscience and Behavior, Barnard College, Columbia University, New York, NY, 10032, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Mark S Ansorge
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA.
- Department of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, 10032, USA.
| |
Collapse
|
29
|
Zeng PY, Tsai YH, Lee CL, Ma YK, Kuo TH. Minimal influence of estrous cycle on studies of female mouse behaviors. Front Mol Neurosci 2023; 16:1146109. [PMID: 37470056 PMCID: PMC10352621 DOI: 10.3389/fnmol.2023.1146109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Sex bias has been an issue in many biomedical fields, especially in neuroscience. In rodent research, many scientists only focused on male animals due to the belief that female estrous cycle gives rise to unacceptable, high levels of variance in the experiments. However, even though female sexual behaviors are well known to be regulated by estrous cycle, which effects on other non-sexual behaviors were not always consistent in previous reports. Recent reviews analyzing published literature even suggested that there is no evidence for larger variation in female than male in several phenotypes. Methods To further investigate the impact of estrous cycle on the variability of female behaviors, we conducted multiple behavioral assays, including the open field test, forced swimming test, and resident-intruder assay to assess anxiety-, depression-like behaviors, as well as social interaction respectively. We compared females in the estrus and diestrus stages across four different mouse strains: C57BL/6, BALB/c, C3H, and DBA/2. Results Our results found no significant difference in most behavioral parameters between females in these two stages. On the other hand, the differences in behaviors among certain strains are relatively consistent in both stages, suggesting a very minimal effect of estrous cycle for detecting the behavioral difference. Last, we compared the behavioral variation between male and female and found very similar variations in most behaviors between the two sexes. Discussion While our study successfully identified behavioral differences among strains and between the sexes, we did not find solid evidence to support the notion that female behaviors are influenced by the estrous cycle. Additionally, we observed similar levels of behavioral variability between males and females. Female mice, therefore, have no reason to be excluded in future behavioral research.
Collapse
Affiliation(s)
- Pei-Yun Zeng
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Ya-Hsuan Tsai
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Lin Lee
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Kai Ma
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
| | - Tsung-Han Kuo
- Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
- Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
30
|
Rodriguez LA, Tran MN, Garcia-Flores R, Pattie EA, Divecha HR, Kim SH, Shin JH, Lee YK, Montoya C, Jaffe AE, Collado-Torres L, Page SC, Martinowich K. TrkB-dependent regulation of molecular signaling across septal cell types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.29.547069. [PMID: 37425939 PMCID: PMC10327212 DOI: 10.1101/2023.06.29.547069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
The lateral septum (LS), a GABAergic structure located in the basal forebrain, is implicated in social behavior, learning and memory. We previously demonstrated that expression of tropomyosin kinase receptor B (TrkB) in LS neurons is required for social novelty recognition. To better understand molecular mechanisms by which TrkB signaling controls behavior, we locally knocked down TrkB in LS and used bulk RNA-sequencing to identify changes in gene expression downstream of TrkB. TrkB knockdown induces upregulation of genes associated with inflammation and immune responses, and downregulation of genes associated with synaptic signaling and plasticity. Next, we generated one of the first atlases of molecular profiles for LS cell types using single nucleus RNA-sequencing (snRNA-seq). We identified markers for the septum broadly, and the LS specifically, as well as for all neuronal cell types. We then investigated whether the differentially expressed genes (DEGs) induced by TrkB knockdown map to specific LS cell types. Enrichment testing identified that downregulated DEGs are broadly expressed across neuronal clusters. Enrichment analyses of these DEGs demonstrated that downregulated genes are uniquely expressed in the LS, and associated with either synaptic plasticity or neurodevelopmental disorders. Upregulated genes are enriched in LS microglia, associated with immune response and inflammation, and linked to both neurodegenerative disease and neuropsychiatric disorders. In addition, many of these genes are implicated in regulating social behaviors. In summary, the findings implicate TrkB signaling in the LS as a critical regulator of gene networks associated with psychiatric disorders that display social deficits, including schizophrenia and autism, and with neurodegenerative diseases, including Alzheimer's.
Collapse
Affiliation(s)
- Lionel A. Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Matthew Nguyen Tran
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Renee Garcia-Flores
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A. Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Joo Heon Shin
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Yong Kyu Lee
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Carly Montoya
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Andrew E. Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leonardo Collado-Torres
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21205, USA
| |
Collapse
|
31
|
Mikami K, Watanabe N, Tochio T, Kimoto K, Akama F, Yamamoto K. Impact of Gut Microbiota on Host Aggression: Potential Applications for Therapeutic Interventions Early in Development. Microorganisms 2023; 11:microorganisms11041008. [PMID: 37110431 PMCID: PMC10141163 DOI: 10.3390/microorganisms11041008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 04/29/2023] Open
Abstract
Aggression in the animal kingdom is a necessary component of life; however, certain forms of aggression, especially in humans, are pathological behaviors that are detrimental to society. Animal models have been used to study a number of factors, including brain morphology, neuropeptides, alcohol consumption, and early life circumstances, to unravel the mechanisms underlying aggression. These animal models have shown validity as experimental models. Moreover, recent studies using mouse, dog, hamster, and drosophila models have indicated that aggression may be affected by the "microbiota-gut-brain axis." Disturbing the gut microbiota of pregnant animals increases aggression in their offspring. In addition, behavioral analyses using germ-free mice have shown that manipulating the intestinal microbiota during early development suppresses aggression. These studies suggest that treating the host gut microbiota during early development is critical. However, few clinical studies have investigated gut-microbiota-targeted treatments with aggression as a primary endpoint. This review aims to clarify the effects of gut microbiota on aggression and discusses the therapeutic potential of regulating human aggression by intervening in gut microbiota.
Collapse
Affiliation(s)
- Katsunaka Mikami
- Department of Psychiatry, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| | - Natsuru Watanabe
- Department of Psychiatry, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| | - Takumi Tochio
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Keitaro Kimoto
- Department of Psychiatry, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| | - Fumiaki Akama
- Department of Psychiatry, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| | - Kenji Yamamoto
- Department of Psychiatry, Tokai University School of Medicine, Isehara 259-1193, Kanagawa, Japan
| |
Collapse
|
32
|
Potegal M, Nordman JC. Non-angry aggressive arousal and angriffsberietschaft: A narrative review of the phenomenology and physiology of proactive/offensive aggression motivation and escalation in people and other animals. Neurosci Biobehav Rev 2023; 147:105110. [PMID: 36822384 DOI: 10.1016/j.neubiorev.2023.105110] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/14/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023]
Abstract
Human aggression typologies largely correspond with those for other animals. While there may be no non-human equivalent of angry reactive aggression, we propose that human proactive aggression is similar to offense in other animals' dominance contests for territory or social status. Like predation/hunting, but unlike defense, offense and proactive aggression are positively reinforcing, involving dopamine release in accumbens. The drive these motivational states provide must suffice to overcome fear associated with initiating risky fights. We term the neural activity motivating proactive aggression "non-angry aggressive arousal", but use "angriffsberietschaft" for offense motivation in other animals to acknowledge possible differences. Temporal variation in angriffsberietschaft partitions fights into bouts; engendering reduced anti-predator vigilance, redirected aggression and motivational over-ride. Increased aggressive arousal drives threat-to-attack transitions, as in verbal-to-physical escalation and beyond that, into hyper-aggression. Proactive aggression and offense involve related neural activity states. Cingulate, insular and prefrontal cortices energize/modulate aggression through a subcortical core containing subnuclei for each aggression type. These proposals will deepen understanding of aggression across taxa, guiding prevention/intervention for human violence.
Collapse
Affiliation(s)
| | - Jacob C Nordman
- Department of Physiology, Southern Illinois University School of Medicine, Carbondale, IL, USA.
| |
Collapse
|
33
|
Sarkar A, Wrangham RW. Evolutionary and neuroendocrine foundations of human aggression. Trends Cogn Sci 2023; 27:468-493. [PMID: 37003880 DOI: 10.1016/j.tics.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 04/03/2023]
Abstract
Humans present a behavioural paradox: they are peaceful in many circumstances, but they are also violent and kill conspecifics at high rates. We describe a social evolutionary theory to resolve this paradox. The theory interprets human aggression as a combination of low propensities for reactive aggression and coercive behaviour and high propensities for some forms of proactive aggression (especially coalitionary proactive aggression). These tendencies are associated with the evolution of groupishness, self-domestication, and social norms. This human aggression profile is expected to demand substantial plasticity in the evolved biological mechanisms responsible for aggression. We discuss the contributions of various social signalling molecules (testosterone, cortisol, oxytocin, vasopressin, serotonin, and dopamine) as the neuroendocrine foundation conferring such plasticity.
Collapse
Affiliation(s)
- Amar Sarkar
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Richard W Wrangham
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| |
Collapse
|
34
|
Fritz M, Soravia SM, Dudeck M, Malli L, Fakhoury M. Neurobiology of Aggression-Review of Recent Findings and Relationship with Alcohol and Trauma. BIOLOGY 2023; 12:biology12030469. [PMID: 36979161 PMCID: PMC10044835 DOI: 10.3390/biology12030469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023]
Abstract
Aggression can be conceptualized as any behavior, physical or verbal, that involves attacking another person or animal with the intent of causing harm, pain or injury. Because of its high prevalence worldwide, aggression has remained a central clinical and public safety issue. Aggression can be caused by several risk factors, including biological and psychological, such as genetics and mental health disorders, and socioeconomic such as education, employment, financial status, and neighborhood. Research over the past few decades has also proposed a link between alcohol consumption and aggressive behaviors. Alcohol consumption can escalate aggressive behavior in humans, often leading to domestic violence or serious crimes. Converging lines of evidence have also shown that trauma and posttraumatic stress disorder (PTSD) could have a tremendous impact on behavior associated with both alcohol use problems and violence. However, although the link between trauma, alcohol, and aggression is well documented, the underlying neurobiological mechanisms and their impact on behavior have not been properly discussed. This article provides an overview of recent advances in understanding the translational neurobiological basis of aggression and its intricate links to alcoholism and trauma, focusing on behavior. It does so by shedding light from several perspectives, including in vivo imaging, genes, receptors, and neurotransmitters and their influence on human and animal behavior.
Collapse
Affiliation(s)
- Michael Fritz
- School of Health and Social Sciences, AKAD University of Applied Sciences, 70191 Stuttgart, Germany
- Department of Forensic Psychiatry and Psychotherapy, Ulm University, BKH Günzburg, Lindenallee 2, 89312 Günzburg, Germany
| | - Sarah-Maria Soravia
- Department of Forensic Psychiatry and Psychotherapy, Ulm University, BKH Günzburg, Lindenallee 2, 89312 Günzburg, Germany
| | - Manuela Dudeck
- Department of Forensic Psychiatry and Psychotherapy, Ulm University, BKH Günzburg, Lindenallee 2, 89312 Günzburg, Germany
| | - Layal Malli
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut P.O. Box 13-5053, Lebanon
| | - Marc Fakhoury
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Beirut P.O. Box 13-5053, Lebanon
| |
Collapse
|
35
|
D1 receptor-expressing neurons in ventral tegmental area alleviate mouse anxiety-like behaviors via glutamatergic projection to lateral septum. Mol Psychiatry 2023; 28:625-638. [PMID: 36195641 PMCID: PMC9531220 DOI: 10.1038/s41380-022-01809-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022]
Abstract
Dopamine (DA) acts as a key regulator in controlling emotion, and dysfunction of DA signal has been implicated in the pathophysiology of some psychiatric disorders, including anxiety. Ventral tegmental area (VTA) is one of main regions with DA-producing neurons. VTA DAergic projections in mesolimbic brain regions play a crucial role in regulating anxiety-like behaviors, however, the function of DA signal within VTA in regulating emotion remains unclear. Here, we observe that pharmacological activation/inhibition of VTA D1 receptors will alleviate/aggravate mouse anxiety-like behaviors, and knockdown of VTA D1 receptor expression also exerts anxiogenic effect. With fluorescence in situ hybridization and electrophysiological recording, we find that D1 receptors are functionally expressed in VTA neurons. Silencing/activating VTA D1 neurons bidirectionally modulate mouse anxiety-like behaviors. Furthermore, knocking down D1 receptors in VTA DA and glutamate neurons elevates anxiety-like state, but in GABA neurons has the opposite effect. In addition, we identify the glutamatergic projection from VTA D1 neurons to lateral septum is mainly responsible for the anxiolytic effect induced by activating VTA D1 neurons. Thus, our study not only characterizes the functional expression of D1 receptors in VTA neurons, but also uncovers the pivotal role of DA signal within VTA in mediating anxiety-like behaviors.
Collapse
|
36
|
Liu D, Hu H, Hong Y, Xiao Q, Tu J. Sugar Beverage Habitation Relieves Chronic Stress-Induced Anxiety-like Behavior but Elicits Compulsive Eating Phenotype via vLS GAD2 Neurons. Int J Mol Sci 2022; 24:ijms24010661. [PMID: 36614104 PMCID: PMC9820526 DOI: 10.3390/ijms24010661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 01/03/2023] Open
Abstract
Chronically stressed individuals are reported to overconsume tasty, palatable foods like sucrose to blunt the psychological and physiological impacts of stress. Negative consequences of high-sugar intake on feeding behavior include increased metabolic disease burdens like obesity. However, the neural basis underlying long-term high-sugar intake-induced overeating during stress is not fully understood. To investigate this question, we used the two-bottle sucrose choice paradigm in mice exposed to chronic unpredictable mild stressors (CUMS) that mimic those of daily life stressors. After 21 days of CUMS paralleled by consecutive sucrose drinking, we explored anxiety-like behavior using the elevated plus maze and open field tests. The normal water-drinking stressed mice displayed more anxiety than the sucrose-drinking stressed mice. Although sucrose-drinking displayed anxiolytic effects, the sucrose-drinking mice exhibited binge eating (chow) and a compulsive eating phenotype. The sucrose-drinking mice also showed a significant body-weight gain compared to the water-drinking control mice during stress. We further found that c-Fos expression was significantly increased in the ventral part of the lateral septum (vLS) of the sucrose-treated stressed mice after compulsive eating. Pharmacogenetic activation of the vLS glutamate decarboxylase 2(GAD2) neurons maintained plain chow intake but induced a compulsive eating phenotype in the naïve GAD2-Cre mice when mice feeding was challenged by flash stimulus, mimicking the negative consequences of excessive sucrose drinking during chronic stress. Further, pharmacogenetic activation of the vLSGAD2 neurons aggravated anxiety of the stressed GAD2-Cre mice but did not alter the basal anxiety level of the naïve ones. These findings indicate the GABAergic neurons within the vLS may be a potential intervention target for anxiety comorbid eating disorders during stress.
Collapse
Affiliation(s)
- Dan Liu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Haohao Hu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuchuan Hong
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Xiao
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jie Tu
- Shenzhen Key Laboratory of Neuroimmunomodulation for Neurological Diseases, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
37
|
Zhukov IS, Karpova IV, Krotova NA, Tissen IY, Demin KA, Shabanov PD, Budygin EA, Kalueff AV, Gainetdinov RR. Enhanced Aggression, Reduced Self-Grooming Behavior and Altered 5-HT Regulation in the Frontal Cortex in Mice Lacking Trace Amine-Associated Receptor 1 (TAAR1). Int J Mol Sci 2022; 23:ijms232214066. [PMID: 36430544 PMCID: PMC9695497 DOI: 10.3390/ijms232214066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
The Trace Amine-Associated Receptor 1 (TAAR1) is one of the six functional receptors belonging to the family of monoamine-related G protein-coupled receptors (TAAR1-TAAR9) found in humans. However, the exact biological mechanisms of TAAR1 central and peripheral action remain to be fully understood. TAAR1 is widely expressed in the prefrontal cortex and several limbic regions, interplaying with the dopamine system to modulate the reward circuitry. Recent clinical trials suggest the efficacy of TAAR1 agonists as potential novel antipsychotic agents. Here, we characterize behavioral and neurochemical phenotypes of TAAR1 knockout mice, focusing on aggression and self-grooming behavior that both strongly depend on the monoaminergic signaling and cortico-striatal and cortico-limbic circuits. Overall, we report increased aggression in these knockout mice in the resident-intruder test, accompanied by reduced self-grooming behavior in the novelty-induced grooming test, and by higher cortical serotonin (5-HT) tissue levels. Further studies are necessary to explore whether TAAR1-based therapies can become potential novel treatments for a wide range of neuropsychiatric disorders associated with aggression.
Collapse
Affiliation(s)
- Ilya S. Zhukov
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Inessa V. Karpova
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Nataliya A. Krotova
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
| | - Ilya Y. Tissen
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Konstantin A. Demin
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
| | - Petr D. Shabanov
- Institute of Experimental Medicine, Acad. Pavlov str. 12, 197376 St. Petersburg, Russia
| | - Evgeny A. Budygin
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Allan V. Kalueff
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, 197341 St. Petersburg, Russia
- Neurobiology Program, Sirius University of Science and Technology, 354340 Sochi, Russia
- Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, 197758 St. Petersburg, Russia
- Neurobiology Laboratory, Ural Federal University, 620002 Yekaterinburg, Russia
- Laboratory of Cell and Molecular Biology and Neurobiology, School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Moscow, Russia
| | - Raul R. Gainetdinov
- Institute of Translational Biomedicine, St. Petersburg State University, University nab. 7/9, 199034 St. Petersburg, Russia
- St. Petersburg University Hospital, St. Petersburg State University, 199034 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
38
|
Transcription Factors as Important Regulators of Changes in Behavior through Domestication of Gray Rats: Quantitative Data from RNA Sequencing. Int J Mol Sci 2022; 23:ijms232012269. [PMID: 36293128 PMCID: PMC9603081 DOI: 10.3390/ijms232012269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/28/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
Studies on hereditary fixation of the tame-behavior phenotype during animal domestication remain relevant and important because they are of both basic research and applied significance. In model animals, gray rats Rattus norvegicus bred for either an enhancement or reduction in defensive response to humans, for the first time, we used high-throughput RNA sequencing to investigate differential expression of genes in tissue samples from the tegmental region of the midbrain in 2-month-old rats showing either tame or aggressive behavior. A total of 42 differentially expressed genes (DEGs; adjusted p-value < 0.01 and fold-change > 2) were identified, with 20 upregulated and 22 downregulated genes in the tissue samples from tame rats compared with aggressive rats. Among them, three genes encoding transcription factors (TFs) were detected: Ascl3 was upregulated, whereas Fos and Fosb were downregulated in tissue samples from the brains of tame rats brain. Other DEGs were annotated as associated with extracellular matrix components, transporter proteins, the neurotransmitter system, signaling molecules, and immune system proteins. We believe that these DEGs encode proteins that constitute a multifactorial system determining the behavior for which the rats have been artificially selected. We demonstrated that several structural subtypes of E-box motifs—known as binding sites for many developmental TFs of the bHLH class, including the ASCL subfamily of TFs—are enriched in the set of promoters of the DEGs downregulated in the tissue samples of tame rats’. Because ASCL3 may act as a repressor on target genes of other developmental TFs of the bHLH class, we hypothesize that the expression of TF gene Ascl3 in tame rats indicates longer neurogenesis (as compared to aggressive rats), which is a sign of neoteny and domestication. Thus, our domestication model shows a new function of TF ASCL3: it may play the most important role in behavioral changes in animals.
Collapse
|
39
|
Meyer D, Athota P, Gowen A, Nguyen NM, Schaal VL, Yelamanchili SV, Pendyala G. Effect of Combined Methamphetamine and Oxycodone Use on the Synaptic Proteome in an In Vitro Model of Polysubstance Use. Genes (Basel) 2022; 13:genes13101816. [PMID: 36292701 PMCID: PMC9601452 DOI: 10.3390/genes13101816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 01/03/2023] Open
Abstract
Polysubstance use (PSU) generally involves the simultaneous use of an opioid along with a stimulant. In recent years, this problem has escalated into a nationwide epidemic. Understanding the mechanisms and effects underlying the interaction between these drugs is essential for the development of treatments for those suffering from addiction. Currently, the effect of PSU on synapses-critical points of contact between neurons-remains poorly understood. Using an in vitro model of primary neurons, we examined the combined effects of the psychostimulant methamphetamine (METH) and the prescription opioid oxycodone (oxy) on the synaptic proteome using quantitative mass-spectrometry-based proteomics. A further ClueGO analysis and Ingenuity Pathway Analysis (IPA) indicated the dysregulation of several molecular functions, biological processes, and pathways associated with neural plasticity and structural development. We identified one key synaptic protein, Striatin-1, which plays a vital role in many of these processes and functions, to be downregulated following METH+oxy treatment. This downregulation of Striatin-1 was further validated by Western blot. Overall, the present study indicates several damaging effects of the combined use of METH and oxy on neural function and warrants further detailed investigation into mechanisms contributing to synaptic dysfunction.
Collapse
Affiliation(s)
- Daniel Meyer
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Pranavi Athota
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Austin Gowen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nghi M. Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Victoria L. Schaal
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sowmya V. Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Genetics, Cell Biology, and Anatomy; University of Nebraska Medical Center, Omaha, NE 68198, USA
- National Strategic Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-559-8690
| |
Collapse
|
40
|
Palavicino-Maggio CB, Sengupta S. The Neuromodulatory Basis of Aggression: Lessons From the Humble Fruit Fly. Front Behav Neurosci 2022; 16:836666. [PMID: 35517573 PMCID: PMC9062135 DOI: 10.3389/fnbeh.2022.836666] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/07/2022] [Indexed: 11/22/2022] Open
Abstract
Aggression is an intrinsic trait that organisms of almost all species, humans included, use to get access to food, shelter, and mating partners. To maximize fitness in the wild, an organism must vary the intensity of aggression toward the same or different stimuli. How much of this variation is genetic and how much is externally induced, is largely unknown but is likely to be a combination of both. Irrespective of the source, one of the principal physiological mechanisms altering the aggression intensity involves neuromodulation. Any change or variation in aggression intensity is most likely governed by a complex interaction of several neuromodulators acting via a meshwork of neural circuits. Resolving aggression-specific neural circuits in a mammalian model has proven challenging due to the highly complex nature of the mammalian brain. In that regard, the fruit fly model Drosophila melanogaster has provided insights into the circuit-driven mechanisms of aggression regulation and its underlying neuromodulatory basis. Despite morphological dissimilarities, the fly brain shares striking similarities with the mammalian brain in genes, neuromodulatory systems, and circuit-organization, making the findings from the fly model extremely valuable for understanding the fundamental circuit logic of human aggression. This review discusses our current understanding of how neuromodulators regulate aggression based on findings from the fruit fly model. We specifically focus on the roles of Serotonin (5-HT), Dopamine (DA), Octopamine (OA), Acetylcholine (ACTH), Sex Peptides (SP), Tachykinin (TK), Neuropeptide F (NPF), and Drosulfakinin (Dsk) in fruit fly male and female aggression.
Collapse
Affiliation(s)
- Caroline B Palavicino-Maggio
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Boston, MA, United States.,Department of Neurobiology, Harvard Medical School, Boston, MA, United States
| | - Saheli Sengupta
- Basic Neuroscience Division, Department of Psychiatry, Harvard Medical School, McLean Hospital, Boston, MA, United States
| |
Collapse
|
41
|
Faure P, Fayad SL, Solié C, Reynolds LM. Social Determinants of Inter-Individual Variability and Vulnerability: The Role of Dopamine. Front Behav Neurosci 2022; 16:836343. [PMID: 35386723 PMCID: PMC8979673 DOI: 10.3389/fnbeh.2022.836343] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
Individuals differ in their traits and preferences, which shape their interactions, their prospects for survival and their susceptibility to diseases. These correlations are well documented, yet the neurophysiological mechanisms underlying the emergence of distinct personalities and their relation to vulnerability to diseases are poorly understood. Social ties, in particular, are thought to be major modulators of personality traits and psychiatric vulnerability, yet the majority of neuroscience studies are performed on rodents in socially impoverished conditions. Rodent micro-society paradigms are therefore key experimental paradigms to understand how social life generates diversity by shaping individual traits. Dopamine circuitry is implicated at the interface between social life experiences, the expression of essential traits, and the emergence of pathologies, thus proving a possible mechanism to link these three concepts at a neuromodulatory level. Evaluating inter-individual variability in automated social testing environments shows great promise for improving our understanding of the link between social life, personality, and precision psychiatry – as well as elucidating the underlying neurophysiological mechanisms.
Collapse
|
42
|
Besnard A, Leroy F. Top-down regulation of motivated behaviors via lateral septum sub-circuits. Mol Psychiatry 2022; 27:3119-3128. [PMID: 35581296 PMCID: PMC7613864 DOI: 10.1038/s41380-022-01599-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022]
Abstract
How does cognition regulate innate behaviors? While the cognitive functions of the cortex have been extensively studied, we know much less about how cognition can regulate innate motivated behaviors to fulfill physiological, safety and social needs. Selection of appropriate motivated behaviors depends on external stimuli and past experiences that helps to scale priorities. With its abundant inputs from neocortical and allocortical regions, the lateral septum (LS) is ideally positioned to integrate perception and experience signals in order to regulate the activity of hypothalamic and midbrain nuclei that control motivated behaviors. In addition, LS receives numerous subcortical modulatory inputs, which represent the animal internal states and also participate in this regulation. In this perspective, we argue that LS sub-circuits regulate distinct motivated behaviors by integrating neural activity from neocortical, allocortical and neuromodulatory inputs. In addition, we propose that lateral inhibition between LS sub-circuits may allow the emergence of functional units that orchestrates competing motivated behaviors.
Collapse
Affiliation(s)
| | - Felix Leroy
- Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, Spain.
| |
Collapse
|