1
|
Ritger AC, Rasheed NM, Padival M, Ferrara NC, Rosenkranz JA. Prior Negative Experience Biases Activity of Medial Amygdala during Interstrain Social Engagement in Male Rats. eNeuro 2024; 11:ENEURO.0288-24.2024. [PMID: 39260890 PMCID: PMC11419602 DOI: 10.1523/eneuro.0288-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024] Open
Abstract
Social recognition is an essential part of social function and often promotes specific social behaviors based on prior experience. Social and defensive behaviors in particular often emerge with prior experiences of familiarity or novelty/stress, respectively. This is also commonly seen in rodents toward same-strain and interstrain conspecifics. Medial amygdala (MeA) activity guides social choice based on age and sex recognition and is sensitive to social experiences. However, little is known about whether the MeA exhibits differential responses based on strain or how this is impacted by experience. Social stress impacts posterior MeA (MeAp) function and can shift measures of social engagement. However, it is unclear how stress impacts MeAp activity and contributes to altered social behavior. The primary goal of this study in adult male Sprague Dawley rats was to determine whether prior stress experience with a different-strain (Long-Evans) rat impacts MeAp responses to same-strain and different-strain conspecifics in parallel with a change in behavior using in vivo fiber photometry. We found that MeAp activity was uniformly activated during social contact with a novel same-strain rat during a three-chamber social preference test following control handling but became biased toward a novel different-strain rat following social stress. Socially stressed rats also showed initially heightened social interaction with novel same-strain rats but showed social avoidance and fragmented social behavior with novel different-strain rats relative to controls. These results indicate that heightened MeAp activity may guide social responses to novel, threatening, rather than non-threatening, social stimuli after stress.
Collapse
Affiliation(s)
- Alexandra C Ritger
- Department of Foundational Sciences and Humanities, Discipline of Neuroscience, Rosalind Franklin University, North Chicago, Illinois 60064
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Nimah M Rasheed
- Department of Foundational Sciences and Humanities, Discipline of Cellular & Molecular Pharmacology, Rosalind Franklin University, North Chicago, Illinois 60064
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Mallika Padival
- Department of Foundational Sciences and Humanities, Discipline of Cellular & Molecular Pharmacology, Rosalind Franklin University, North Chicago, Illinois 60064
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University, North Chicago, Illinois 60064
| | - Nicole C Ferrara
- Department of Foundational Sciences and Humanities, Discipline of Cellular & Molecular Pharmacology, Rosalind Franklin University, North Chicago, Illinois 60064
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University, North Chicago, Illinois 60064
| | - J Amiel Rosenkranz
- Department of Foundational Sciences and Humanities, Discipline of Cellular & Molecular Pharmacology, Rosalind Franklin University, North Chicago, Illinois 60064
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Rosalind Franklin University, North Chicago, Illinois 60064
| |
Collapse
|
2
|
Wang L, Clark EA, Hanratty L, Koblan KS, Foley A, Dedic N, Bristow LJ. TAAR1 and 5-HT 1B receptor agonists attenuate autism-like irritability and aggression in rats prenatally exposed to valproic acid. Pharmacol Biochem Behav 2024; 245:173862. [PMID: 39197535 DOI: 10.1016/j.pbb.2024.173862] [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: 05/30/2024] [Revised: 08/02/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Despite the rising prevalence of autism spectrum disorder (ASD), there remains a significant unmet need for pharmacotherapies addressing its core and associative symptoms. While some atypical antipsychotics have been approved for managing associated irritability and aggression, their use is constrained by substantial side effects. This study aimed firstly to develop behavioral measures to explore frustration, irritability and aggression phenotypes in the rat prenatal valproic acid (VPA) model of ASD. Additionally, we investigated the potential of two novel mechanisms, 5-HT1B and TAAR1 agonism, to alleviate these behaviors. Male offspring exposed to prenatal VPA were trained to achieve stable performance on a cued operant task, followed by pharmacological assessment in an operant frustration test, bottle brush test and resident intruder test. VPA exposed rats demonstrated behaviors indicative of frustration and irritability, as well as increased aggression compared to controls. The irritability-like behavior and aggression were further exacerbated in animals previously experiencing a frustrative event during the operant test. Single administration of the 5-HT1B agonist CP-94253 or TAAR1 agonist RO5263397 attenuated the frustration-like behavior compared to vehicle. Additionally, both agonists reduced irritability-like behavior under both normal and frustrative conditions. While CP-94253 reduced aggression in the resident intruder test under both conditions, RO5263397 only produced effects in rats that previously experienced a frustrative event. Our study describes previously uncharacterized phenotypes of frustration, irritability, and aggression in the rat prenatal VPA model of ASD. Administration of selective TAAR1 or 5-HT1B receptor agonists alleviated these deficits, warranting further exploration of both targets in ASD treatment.
Collapse
Affiliation(s)
- Lien Wang
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | - Erin A Clark
- Sumitomo Pharma America, Inc., Marlborough, MA, USA
| | | | | | | | - Nina Dedic
- Sumitomo Pharma America, Inc., Marlborough, MA, USA.
| | | |
Collapse
|
3
|
Grammer J, Valles R, Bowles A, Zelikowsky M. SAUSI: a novel assay for measuring social anxiety and motivation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594023. [PMID: 38798428 PMCID: PMC11118329 DOI: 10.1101/2024.05.13.594023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Social anxiety is one of the most prevalent mental health disorders, though the underlying neurobiology is poorly understood. Progress in understanding the etiology of social anxiety has been hindered by the lack of comprehensive tools to assess social anxiety in model systems. Here, we created a new behavioral task - Selective Access to Unrestricted Social Interaction (SAUSI), which combines elements of social motivation, hesitancy, decision-making, and free interaction to enable the wholistic assessment of social anxiety-like behaviors in mice. Using this novel assay, we found that social isolation-induced social anxiety-like behaviors in female mice are largely driven by increases in social fear, social hesitancy, and altered ultrasonic vocalizations. Deep learning analyses were able to computationally identify a unique behavioral footprint underlying the state produced by social isolation, demonstrating the compatibility of modern computational approaches with SAUSI. Finally, we compared the results of SAUSI to traditionally social assays including the 3-chamber sociability assay and the resident intruder task. This revealed that behavioral changes induced by isolation were highly context dependent, and that while fragments of social anxiety measured in SAUSI were replicable across other tasks, a wholistic assessment was not obtainable from these alternative assays. Our findings debut a novel task for the behavioral toolbox - one which overcomes limitations of previous assays, allowing for both social choice as well as free interaction, and offers a new approach for assessing social anxiety in rodents.
Collapse
Affiliation(s)
- Jordan Grammer
- Department of Neurobiology, University of Utah, United States
| | - Rene Valles
- Department of Neurobiology, University of Utah, United States
| | - Alexis Bowles
- Department of Neurobiology, University of Utah, United States
| | | |
Collapse
|
4
|
Yin X, Zhao Y, Wang S, Feng H, He X, Li X, Liu X, Lu H, Wen D, Shi Y, Shi H. Postweaning stress affects behavior, brain and gut microbiota of adolescent mice in a sex-dependent manner. Neuropharmacology 2024; 248:109869. [PMID: 38354850 DOI: 10.1016/j.neuropharm.2024.109869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 02/16/2024]
Abstract
Aggression is an instinctive behavior that has been reported to be influenced by early-life stress. However, the potential effects of acute stress during the postweaning period, a key stage for brain development, on defensive aggression and the associated mechanism remain poorly understood. In the present study, aggressive behaviors were evaluated in adolescent mice exposed to postweaning stress. Serum corticosterone and testosterone levels, neural dendritic spine density, and gut microbiota composition were determined to identify the underlying mechanism. Behavioral analysis showed that postweaning stress reduced locomotor activity in mice and decreased defensive aggression in male mice. ELISA results showed that postweaning stress reduced serum testosterone levels in female mice. Golgi staining analysis demonstrated that postweaning stress decreased neural dendritic spine density in the medial prefrontal cortex of male mice. 16S rRNA sequencing results indicated that postweaning stress altered the composition of the gut microbiota in male mice. Combined, these results suggested that postweaning stress alters defensive aggression in male mice, which may be due to changes in neuronal structure as well as gut microbiota composition. Our findings highlight the long-lasting and sex-dependent effects of early-life experience on behaviors.
Collapse
Affiliation(s)
- Xueyong Yin
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ye Zhao
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuang Wang
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hao Feng
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xinyue He
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xincheng Li
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Xiaoyu Liu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Hengtai Lu
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang, 050017, China
| | - Yun Shi
- Neuroscience Research Center, Institute of Medical and Health Science, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Neurophysiology, Hebei Medical 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 Neurophysiology, Hebei Medical University, Shijiazhuang, 050017, China; Hebei Key Laboratory of Forensic Medicine, Hebei Province, Shijiazhuang, 050017, China; Nursing School, Hebei Medical University, Shijiazhuang, 050031, China.
| |
Collapse
|
5
|
Abstract
Norms permeate human life. Most of people's activities can be characterized by rules about what is appropriate, allowed, required, or forbidden-rules that are crucial in making people hyper-cooperative animals. In this article, I examine the current cognitive-evolutionary account of "norm psychology" and propose an alternative that is better supported by evidence and better placed to promote interdisciplinary dialogue. The incumbent theory focuses on rules and claims that humans genetically inherit cognitive and motivational mechanisms specialized for processing these rules. The cultural-evolutionary alternative defines normativity in relation to behavior-compliance, enforcement, and commentary-and suggests that it depends on implicit and explicit processes. The implicit processes are genetically inherited and domain-general; rather than being specialized for normativity, they do many jobs in many species. The explicit processes are culturally inherited and domain-specific; they are constructed from mentalizing and reasoning by social interaction in childhood. The cultural-evolutionary, or "cognitive gadget," perspective suggests that people alive today-parents, educators, elders, politicians, lawyers-have more responsibility for sustaining normativity than the nativist view implies. People's actions not only shape and transmit the rules, but they also create in each new generation mental processes that can grasp the rules and put them into action.
Collapse
Affiliation(s)
- Cecilia Heyes
- Department of Experimental Psychology & All Souls College, University of Oxford
| |
Collapse
|
6
|
Morishita M, Kobayashi K, Mitsuzuka M, Takagi R, Ono K, Momma R, Tsuneoka Y, Horio S, Tsukahara S. Two-Step Actions of Testicular Androgens in the Organization of a Male-Specific Neural Pathway from the Medial Preoptic Area to the Ventral Tegmental Area for Modulating Sexually Motivated Behavior. J Neurosci 2023; 43:7322-7336. [PMID: 37722849 PMCID: PMC10621776 DOI: 10.1523/jneurosci.0361-23.2023] [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: 02/27/2023] [Revised: 08/16/2023] [Accepted: 09/13/2023] [Indexed: 09/20/2023] Open
Abstract
The medial preoptic area (MPOA) is a sexually dimorphic region of the brain that regulates social behaviors. The sexually dimorphic nucleus (SDN) of the MPOA has been studied to understand sexual dimorphism, although the anatomy and physiology of the SDN is not fully understood. Here, we characterized SDN neurons that contribute to sexual dimorphism and investigated the mechanisms underlying the emergence of such neurons and their roles in social behaviors. A target-specific neuroanatomical study using transgenic mice expressing Cre recombinase under the control of Calb1, a gene expressed abundantly in the SDN, revealed that SDN neurons are divided into two subpopulations, GABA neurons projecting to the ventral tegmental area (VTA), where they link to the dopamine system (CalbVTA neurons), and GABA neurons that extend axons in the MPOA or project to neighboring regions (CalbnonVTA neurons). CalbVTA neurons were abundant in males, but were scarce or absent in females. There was no difference in the number of CalbnonVTA neurons between sexes. Additionally, we found that emergence of CalbVTA neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. Chemogenetic analyses of CalbVTA neurons indicated a role in modulating sexual motivation in males. Knockdown of Calb1 in the MPOA reduced the intromission required for males to complete copulation. These findings provide strong evidence that a male-specific neural pathway from the MPOA to the VTA is organized by the two-step actions of testicular androgens for the modulation of sexually motivated behavior.SIGNIFICANCE STATEMENT The MPOA is a sexually dimorphic region of the brain that regulates social behaviors, although its sexual dimorphism is not fully understood. Here, we describe a population of MPOA neurons that contribute to the sexual dimorphism. These neurons only exist in masculinized brains, and they project their axons to the ventral tegmental area, where they link to the dopamine system. Emergence of such neurons requires two testicular androgen actions that occur first in the postnatal period and second in the peripubertal period. These MPOA neurons endow masculinized brains with a neural pathway from the MPOA to the ventral tegmental area and modulate sexually motivated behavior in males.
Collapse
Affiliation(s)
- Masahiro Morishita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Kaito Kobayashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Moeri Mitsuzuka
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Ryo Takagi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Kota Ono
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Rami Momma
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Yousuke Tsuneoka
- Department of Anatomy, Faculty of Medicine, Toho University, Tokyo 43-8540, Japan
| | - Shuhei Horio
- Division of Endocrinology and Metabolism, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| |
Collapse
|
7
|
Börchers S, Carl J, Schormair K, Krieger JP, Asker M, Edvardsson CE, Jerlhag E, Skibicka KP. An appetite for aggressive behavior? Female rats, too, derive reward from winning aggressive interactions. Transl Psychiatry 2023; 13:331. [PMID: 37891191 PMCID: PMC10611704 DOI: 10.1038/s41398-023-02608-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
While aggression is an adaptive behavior mostly triggered by competition for resources, it can also in and of itself be rewarding. Based on the common notion that female rats are not aggressive, much of aggression research has been centered around males, leading to a gap in the understanding of the female aggression neurobiology. Therefore, we asked whether intact virgin female rats experience reward from an aggressive interaction and assessed aggression seeking behavior in rats of both sexes. To validate the involvement of reward signaling, we measured mesolimbic dopamine turnover and determined the necessity of dopamine signaling for expression of aggression-seeking. Together our data indicate that female rats exhibit aggressive behavior outside of maternal context, experience winning aggressive behaviors as rewarding, and do so to a similar extent as male rats and in a dopamine-dependent manner.
Collapse
Affiliation(s)
- Stina Börchers
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Jil Carl
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Katharina Schormair
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Jean-Philippe Krieger
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Mohammed Asker
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Christian E Edvardsson
- Department of Pharmacology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Elisabeth Jerlhag
- Department of Pharmacology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Karolina P Skibicka
- Department of Physiology, Institute for Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
- Department of Nutritional Sciences and Huck Institutes, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
8
|
Reinhardt PR, Theis CDC, Juckel G, Freund N. Rodent models for mood disorders - understanding molecular changes by investigating social behavior. Biol Chem 2023; 404:939-950. [PMID: 37632729 DOI: 10.1515/hsz-2023-0190] [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: 04/18/2023] [Accepted: 08/14/2023] [Indexed: 08/28/2023]
Abstract
Mood disorders, including depressive and bipolar disorders, are the group of psychiatric disorders with the highest prevalence and disease burden. However, their pathophysiology remains poorly understood. Animal models are an extremely useful tool for the investigation of molecular mechanisms underlying these disorders. For psychiatric symptom assessment in animals, a meaningful behavioral phenotype is needed. Social behaviors constitute naturally occurring complex behaviors in rodents and can therefore serve as such a phenotype, contributing to insights into disorder related molecular changes. In this narrative review, we give a fundamental overview of social behaviors in laboratory rodents, as well as their underlying neuronal mechanisms and their assessment. Relevant behavioral and molecular changes in models for mood disorders are presented and an outlook on promising future directions is given.
Collapse
Affiliation(s)
- Patrick R Reinhardt
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
- International Graduate School of Neuroscience, Ruhr-University Bochum, D-44801 Bochum, Germany
| | - Candy D C Theis
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| | - Georg Juckel
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| | - Nadja Freund
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL-University Hospital, Ruhr-University Bochum, D-44791 Bochum, Germany
| |
Collapse
|
9
|
Scott R, Aubry A, Cuttoli RDD, Rachel FF, Lyonna P, Cathomas F, Burnett C, Yang Y, Yuan C, Lablanca A, Chan K, Lin HY, Froemke R, Li L. A critical role for cortical amygdala circuitry in shaping social encounters. RESEARCH SQUARE 2023:rs.3.rs-3015820. [PMID: 37461537 PMCID: PMC10350173 DOI: 10.21203/rs.3.rs-3015820/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Aggression is an evolutionarily conserved behavior that controls social hierarchies and protects valuable resources like mates, food, and territory. In mice, aggressive behaviour can be broken down into an appetitive phase, which involves approach and investigation, and a consummatory phase, which involves biting, kicking, and wrestling. By performing an unsupervised weighted correlation network analysis on whole-brain c-Fos expression, we identified a cluster of brain regions including hypothalamic and amygdalar sub-regions and olfactory cortical regions highly co-activated in male, but not female aggressors (AGG). The posterolateral cortical amygdala (COApl), an extended olfactory structure, was found to be a hub region based on the number and strength of correlations with other regions in the cluster. Our data further show that estrogen receptor 1 (ESR1)-expressing cells in the COApl exhibit increased activity during attack behaviour, and during bouts of investigation which precede an attack, in male mice only. Chemogenetic or optogenetic inhibition of COApl ESR1 cells in AGG males reduces aggression and increases pro-social investigation without affecting social reward/reinforcement behavior. We further confirmed that COApl ESR1 projections to the ventrolateral portion of the ventromedial hypothalamus and central amygdala are necessary for these behaviours. Collectively, these data suggest that in aggressive males, COApl ESR1 cells respond specifically to social stimuli, thereby enhancing their salience and promoting attack behaviour.
Collapse
Affiliation(s)
| | | | | | | | | | | | - C Burnett
- Icahn School of Medicine at Mount Sinai
| | | | | | | | | | | | | | - Long Li
- Icahn School of Medicine at Mount Sinai
| |
Collapse
|
10
|
Kinoshita M, Okamoto H. Acetylcholine potentiates glutamate transmission from the habenula to the interpeduncular nucleus in losers of social conflict. Curr Biol 2023:S0960-9822(23)00445-1. [PMID: 37105168 DOI: 10.1016/j.cub.2023.03.087] [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: 12/07/2022] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023]
Abstract
Switching behaviors from aggression to submission in losers at the end of conspecific social fighting is essential to avoid serious injury or death. We have previously shown that the experience of defeat induces a loser-specific potentiation in the habenula (Hb)-interpeduncular nucleus (IPN) and show here that this is induced by acetylcholine. Calcium imaging and electrophysiological recording using acute brain slices from winners and losers of fighting behavior in zebrafish revealed that the ventral IPN (vIPN) dominates over the dorsal IPN in the neural response to Hb stimulation in losers. We also show that GluA1 α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunits on the postsynaptic membrane increased in the vIPN of losers. Furthermore, these loser-specific neural properties disappeared in the presence of an α7 nicotinic acetylcholine receptor (nAChR) antagonist and, conversely, were induced in brain slices of winners treated with α7 nAChR agonists. These data suggest that acetylcholine released from Hb terminals in the vIPN induces activation of α7 nAChR followed by an increase in postsynaptic membrane GluA1. This results in an increase in active synapses on postsynaptic neurons, resulting in the potentiation of neurotransmissions to the vIPN. This acetylcholine-induced neuromodulation could be the neural foundation for behavioral switching in losers. Our results could increase our understanding of the mechanisms of various mood disorders such as social anxiety disorder and social withdrawal.
Collapse
Affiliation(s)
- Masae Kinoshita
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; RIKEN CBS-Kao Collaboration Center, Saitama 351-0198, Japan.
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
Moskaliuk VS, Kozhemyakina RV, Khomenko TM, Volcho KP, Salakhutdinov NF, Kulikov AV, Naumenko VS, Kulikova EA. On Associations between Fear-Induced Aggression, Bdnf Transcripts, and Serotonin Receptors in the Brains of Norway Rats: An Influence of Antiaggressive Drug TC-2153. Int J Mol Sci 2023; 24:ijms24020983. [PMID: 36674499 PMCID: PMC9867021 DOI: 10.3390/ijms24020983] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/07/2023] Open
Abstract
The Bdnf (brain-derived neurotrophic factor) gene contains eight regulatory exons (I-VIII) alternatively spliced to the protein-coding exon IX. Only exons I, II, IV, and VI are relatively well studied. The BDNF system and brain serotonergic system are tightly interconnected and associated with aggression. The benzopentathiepine TC-2153 affects both systems and exerts antiaggressive action. Our aim was to evaluate the effects of TC-2153 on the Bdnf exons I-IX's expressions and serotonin receptors' mRNA levels in the brain of rats featuring high aggression toward humans (aggressive) or its absence (tame). Aggressive and tame adult male rats were treated once with vehicle or 10 or 20 mg/kg of TC-2153. mRNA was quantified in the cortex, hippocampus, hypothalamus, and midbrain with real-time PCR. Selective breeding for high aggression or its absence affected the serotonin receptors' and Bdnf exons' transcripts differentially, depending on the genotype (strain) and brain region. TC-2153 had comprehensive effects on the Bdnf exons' expressions. The main trend was downregulation in the hypothalamus and midbrain. TC-2153 increased 5-HT1B receptor hypothalamusc mRNA expression. For the first time, an influence of TC-2153 on the expressions of Bdnf regulatory exons and the 5-HT1B receptor was shown, as was an association between Bdnf regulatory exons and fear-induced aggression involving genetic predisposition.
Collapse
Affiliation(s)
- Vitalii S. Moskaliuk
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Rimma V. Kozhemyakina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Tatyana M. Khomenko
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, SB RAS, 9 Akad. Lavrentieva Ave., 630090 Novosibirsk, Russia
| | - Konstantin P. Volcho
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, SB RAS, 9 Akad. Lavrentieva Ave., 630090 Novosibirsk, Russia
| | - Nariman F. Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, SB RAS, 9 Akad. Lavrentieva Ave., 630090 Novosibirsk, Russia
| | - Alexander V. Kulikov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Vladimir S. Naumenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentyeva Ave., 630090 Novosibirsk, Russia
| | - Elizabeth A. Kulikova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), 10 Akad. Lavrentyeva Ave., 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
14
|
Smagin DA, Galyamina AG, Kovalenko IL, Kudryavtseva NN. Altered Expression of Genes Associated with Major Neurotransmitter Systems in the Reward-Related Brain Regions of Mice with Positive Fighting Experience. Int J Mol Sci 2022; 23:13644. [PMID: 36362437 PMCID: PMC9655062 DOI: 10.3390/ijms232113644] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
The main neurotransmitters in the brain-dopamine, γ-aminobutyric acid (GABA), glutamate, and opioids-are recognized to be the most important for the regulation of aggression and addiction. The aim of this work was to study differentially expressed genes (DEGs) in the main reward-related brain regions, including the ventral tegmental area (VTA), dorsal striatum (STR), ventral striatum (nucleus accumbens, NAcc), prefrontal cortex (PFC), and midbrain raphe nuclei (MRNs), in male mice with 20-day positive fighting experience in daily agonistic interactions. Expression of opioidergic, catecholaminergic, glutamatergic, and GABAergic genes was analyzed to confirm or refute the influence of repeated positive fighting experience on the development of "addiction-like" signs shown in our previous studies. High-throughput RNA sequencing was performed to identify differentially expressed genes in the brain regions of chronically aggressive mice. In the aggressive mice, upregulation of opioidergic genes was shown (Oprk1 in VTA, Pdyn in NAcc, Penk in PFC, and Oprd1 in MRNs and PFC), as was downregulation of genes Opcml and Oprk1 in STR and Pomc in VTA and NAcc. Upregulation of catecholaminergic genes in VTA (Ddc and Slc6a2) and in NAcc (Th and Drd2) and downregulation of some differentially expressed genes in MRNs (Th, Ddc, Dbh, Drd2, Slc18a2, and Sncg) and in VTA (Adra2c, Sncg, and Sncb) were also documented. The expression of GABAergic and glutamatergic genes that participate in drug addiction changed in all brain regions. According to literature data, the proteins encoded by genes Drd2, Oprk1, Oprd1, Pdyn, Penk, and Pomc are directly involved in drug addiction in humans. Thus, our results confirm our earlier claim about the formation of addiction-like signs following repeated positive fighting experience in mice, as shown previously in our biobehavioral studies.
Collapse
Affiliation(s)
| | | | | | - Natalia N. Kudryavtseva
- Neuropathology Modeling Laboratory, Neurogenetics of Social Behavior Sector, FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| |
Collapse
|
15
|
Activation of glucagon-like peptide-1 receptors reduces the acquisition of aggression-like behaviors in male mice. Transl Psychiatry 2022; 12:445. [PMID: 36229445 PMCID: PMC9561171 DOI: 10.1038/s41398-022-02209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/21/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Aggression is a complex social behavior, which is provoked in the defense of limited resources including food and mates. Recent advances show that the gut-brain hormone ghrelin modulates aggressive behaviors. As the gut-brain hormone glucagon-like peptide-1 (GLP-1) reduces food intake and sexual behaviors its potential role in aggressive behaviors is likely. Therefore, we investigated a tentative link between GLP-1 and aggressive behaviors by combining preclinical and human genetic-association studies. The influence of acute or repeated injections of a GLP-1 receptor (GLP-1R) agonist, exendin-4 (Ex4), on aggressive behaviors was assessed in male mice exposed to the resident-intruder paradigm. Besides, possible mechanisms participating in the ability of Ex4 to reduce aggressive behaviors were evaluated. Associations of polymorphisms in GLP-1R genes and overt aggression in males of the CATSS cohort were assessed. In male mice, repeated, but not acute, Ex4 treatment dose-dependently reduced aggressive behaviors. Neurochemical and western blot studies further revealed that putative serotonergic and noradrenergic signaling in nucleus accumbens, specifically the shell compartment, may participate in the interaction between Ex4 and aggression. As high-fat diet (HFD) impairs the responsiveness to GLP-1 on various behaviors the possibility that HFD blunts the ability of Ex4 to reduce aggressive behaviors was explored. Indeed, the levels of aggression was similar in vehicle and Ex4 treated mice consuming HFD. In humans, there were no associations between polymorphisms of the GLP-1R genes and overt aggression. Overall, GLP-1 signaling suppresses acquisition of aggressive behaviors via central neurotransmission and additional studies exploring this link are warranted.
Collapse
|
16
|
Baselmans B, Hammerschlag AR, Noordijk S, Ip H, van der Zee M, de Geus E, Abdellaoui A, Treur JL, van ’t Ent D. The Genetic and Neural Substrates of Externalizing Behavior. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 2:389-399. [PMID: 36324656 PMCID: PMC9616240 DOI: 10.1016/j.bpsgos.2021.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
Background To gain more insight into the biological factors that mediate vulnerability to display externalizing behaviors, we leveraged genome-wide association study summary statistics on 13 externalizing phenotypes. Methods After data classification based on genetic resemblance, we performed multivariate genome-wide association meta-analyses and conducted extensive bioinformatic analyses, including genetic correlation assessment with other traits, Mendelian randomization, and gene set and gene expression analyses. Results The genetic data could be categorized into disruptive behavior (DB) and risk-taking behavior (RTB) factors, and subsequent genome-wide association meta-analyses provided association statistics for DB and RTB (N eff = 523,150 and 1,506,537, respectively), yielding 50 and 257 independent genetic signals. The statistics of DB, much more than RTB, signaled genetic predisposition to adverse cognitive, mental health, and personality outcomes. We found evidence for bidirectional causal influences between DB and substance use behaviors. Gene set analyses implicated contributions of neuronal cell development (DB/RTB) and synapse formation and transcription (RTB) mechanisms. Gene-brain mapping confirmed involvement of the amygdala and hypothalamus and highlighted other candidate regions (cerebellar dentate, cuneiform nucleus, claustrum, paracentral cortex). At the cell-type level, we noted enrichment of glutamatergic neurons for DB and RTB. Conclusions This bottom-up, data-driven study provides new insights into the genetic signals of externalizing behaviors and indicates that commonalities in genetic architecture contribute to the frequent co-occurrence of different DBs and different RTBs, respectively. Bioinformatic analyses supported the DB versus RTB categorization and indicated relevant biological mechanisms. Generally similar gene-brain mappings indicate that neuroanatomical differences, if any, escaped the resolution of our methods.
Collapse
Affiliation(s)
- Bart Baselmans
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Anke R. Hammerschlag
- Child Health Research Centre, The University of Queensland, Brisbane, Queensland, Australia
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Stephany Noordijk
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Hill Ip
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Matthijs van der Zee
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Eco de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Abdel Abdellaoui
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jorien L. Treur
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Dennis van ’t Ent
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Amsterdam Public Health research institute, Amsterdam University Medical Centre, Amsterdam, the Netherlands
- Address correspondence to Dennis van ’t Ent, Ph.D.
| |
Collapse
|
17
|
Padilla-Coreano N, Tye KM, Zelikowsky M. Dynamic influences on the neural encoding of social valence. Nat Rev Neurosci 2022; 23:535-550. [PMID: 35831442 PMCID: PMC9997616 DOI: 10.1038/s41583-022-00609-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 11/09/2022]
Abstract
Social signals can serve as potent emotional triggers with powerful impacts on processes from cognition to valence processing. How are social signals dynamically and flexibly associated with positive or negative valence? How do our past social experiences and present social standing shape our motivation to seek or avoid social contact? We discuss a model in which social attributes, social history, social memory, social rank and social isolation can flexibly influence valence assignment to social stimuli, termed here as 'social valence'. We emphasize how the brain encodes each of these four factors and highlight the neural circuits and mechanisms that play a part in the perception of social attributes, social memory and social rank, as well as how these factors affect valence systems associated with social stimuli. We highlight the impact of social isolation, dissecting the neural and behavioural mechanisms that mediate the effects of acute versus prolonged periods of social isolation. Importantly, we discuss conceptual models that may account for the potential shift in valence of social stimuli from positive to negative as the period of isolation extends in time. Collectively, this Review identifies factors that control the formation and attribution of social valence - integrating diverse areas of research and emphasizing their unique contributions to the categorization of social stimuli as positive or negative.
Collapse
Affiliation(s)
- Nancy Padilla-Coreano
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Kay M Tye
- HHMI-Salk Institute for Biological Studies, La Jolla, CA, USA.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
18
|
Aldhshan MS, Mizuno TM. Effect of environmental enrichment on aggression and the expression of brain-derived neurotrophic factor transcript variants in group-housed male mice. Behav Brain Res 2022; 433:113986. [DOI: 10.1016/j.bbr.2022.113986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/20/2022] [Accepted: 06/28/2022] [Indexed: 11/02/2022]
|
19
|
Börchers S, Krieger JP, Asker M, Maric I, Skibicka KP. Commonly-used rodent tests of anxiety-like behavior lack predictive validity for human sex differences. Psychoneuroendocrinology 2022; 141:105733. [PMID: 35367714 DOI: 10.1016/j.psyneuen.2022.105733] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 01/07/2023]
Abstract
Women are more likely to develop an anxiety disorder than men. Yet, preclinical models of anxiety were largely developed in male rodents, with poorly understood predictive validity for sex differences. Here, we investigate whether commonly-used anxiety-like behavior tests, elevated plus maze (EPM) and open field (OF), represent the human sex difference in adult Sprague-Dawley rats. When interpreted by EPM or OF, female rats displayed less anxiety-like behavior compared to males, as they spent twice as much time in the open arms of the EPM or the center of the OF compared to males. However, they also displayed vastly different levels of locomotor activity, possibly confounding interpretation of these locomotion-dependent tests. To exclude locomotion from the assessment, the acoustic startle response (ASR) test was used. When interpreted by the ASR test, females displayed more anxiety-like behavior compared to males, as indicated by a nearly two-fold higher startle amplitude. The observed sex differences were not driven by gonadal steroids. Overall, all but one of the tests fail to mirror the sex difference in anxiety reported in humans. Our findings suggest that the ASR might be a better fit in modelling female anxiety-like behavior.
Collapse
Affiliation(s)
- Stina Börchers
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - Jean-Philippe Krieger
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - Mohammed Asker
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden
| | - Ivana Maric
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA
| | - Karolina P Skibicka
- Institute for Neuroscience and Physiology, University of Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Sweden; Department of Nutritional Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
20
|
Wang ZJ, Shwani T, Liu J, Zhong P, Yang F, Schatz K, Zhang F, Pralle A, Yan Z. Molecular and cellular mechanisms for differential effects of chronic social isolation stress in males and females. Mol Psychiatry 2022; 27:3056-3068. [PMID: 35449296 PMCID: PMC9615910 DOI: 10.1038/s41380-022-01574-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/06/2022] [Accepted: 04/08/2022] [Indexed: 12/21/2022]
Abstract
Chronic social isolation stress during adolescence induces susceptibility for neuropsychiatric disorders. Here we show that 5-week post-weaning isolation stress induces sex-specific behavioral abnormalities and neuronal activity changes in the prefrontal cortex (PFC), basal lateral amygdala (BLA), and ventral tegmental area (VTA). Chemogenetic manipulation, optogenetic recording, and in vivo calcium imaging identify that the PFC to BLA pathway is causally linked to heightened aggression in stressed males, and the PFC to VTA pathway is causally linked to social withdrawal in stressed females. Isolation stress induces genome-wide transcriptional alterations in a region-specific manner. Particularly, the upregulated genes in BLA of stressed males are under the control of activated transcription factor CREB, and CREB inhibition in BLA normalizes gene expression and reverses aggressive behaviors. On the other hand, neuropeptide Hcrt (Hypocretin/Orexin) is among the top-ranking downregulated genes in VTA of stressed females, and Orexin-A treatment rescues social withdrawal. These results have revealed molecular mechanisms and potential therapeutic targets for stress-related mental illness.
Collapse
Affiliation(s)
- Zi-Jun Wang
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Treefa Shwani
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Junting Liu
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Fengwei Yang
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Kelcie Schatz
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Freddy Zhang
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Arnd Pralle
- Department of Physics, College of Arts and Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
21
|
Grinberg M, Levin R, Neuman H, Ziv O, Turjeman S, Gamliel G, Nosenko R, Koren O. Antibiotics increase aggression behavior and aggression-related pheromones and receptors in Drosophila melanogaster. iScience 2022; 25:104371. [PMID: 35620429 PMCID: PMC9127605 DOI: 10.1016/j.isci.2022.104371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/07/2022] [Accepted: 05/04/2022] [Indexed: 11/05/2022] Open
Abstract
Aggression is a behavior common in most species; it is controlled by internal and external drivers, including hormones, environmental cues, and social interactions, and underlying pathways are understood in a broad range of species. To date, though, effects of gut microbiota on aggression in the context of gut-brain communication and social behavior have not been completely elucidated. We examine how manipulation of Drosophila melanogaster microbiota affects aggression as well as the pathways that underlie the behavior in this species. Male flies treated with antibiotics exhibited significantly more aggressive behaviors. Furthermore, they had higher levels of cVA and (Z)-9 Tricosene, pheromones associated with aggression in flies, as well as higher expression of the relevant pheromone receptors and transporters OR67d, OR83b, GR32a, and LUSH. These findings suggest that aggressive behavior is, at least in part, mediated by bacterial species in flies. Aggression increases in flies that lack a microbiome Monocolonization with specific bacteria can mediate this effect We observed differences in aggression-related pheromone expression levels
Collapse
|
22
|
Baleisyte A, Schneggenburger R, Kochubey O. Stimulation of medial amygdala GABA neurons with kinetically different channelrhodopsins yields opposite behavioral outcomes. Cell Rep 2022; 39:110850. [PMID: 35613578 DOI: 10.1016/j.celrep.2022.110850] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/20/2021] [Accepted: 04/30/2022] [Indexed: 12/11/2022] Open
Abstract
The medial amygdala (MeA) receives pheromone information about conspecifics and has crucial functions in social behaviors. A previous study showed that activation of GABA neurons in the postero-dorsal MeA (MeApd) with channelrhodopsin-2H134R (ChR2) stimulates inter-male aggression. When performing these experiments using the faster channelrhodopsinH134R,E123T (ChETA), we find the opposite behavioral outcome. A systematic comparison between the two channelrhodopsin variants reveals that optogenetic activation of MeApd GABA neurons with ChETA suppresses aggression, whereas activation under ChR2 increases aggression. Although the mechanism for this paradoxical difference is not understood, we observe that activation of MeApd GABA neurons with ChR2 causes larger plateau depolarizations, smaller action potentials, and larger local inhibition than with ChETA. Thus, the channelrhodopsin variant used for in vivo optogenetic experiments can radically influence the behavioral outcome. Future work should continue to study the role of specific sub-populations of MeApd GABA neurons in aggression control.
Collapse
Affiliation(s)
- Aiste Baleisyte
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Ralf Schneggenburger
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Olexiy Kochubey
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
23
|
Ferreira-Fernandes E, Peça J. The Neural Circuit Architecture of Social Hierarchy in Rodents and Primates. Front Cell Neurosci 2022; 16:874310. [PMID: 35634473 PMCID: PMC9133341 DOI: 10.3389/fncel.2022.874310] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/29/2022] [Indexed: 11/13/2022] Open
Abstract
Social status is recognized as a major determinant of social behavior and health among animals; however, the neural circuits supporting the formation and navigation of social hierarchies remain under extensive research. Available evidence suggests the prefrontal cortex is a keystone in this circuit, but upstream and downstream candidates are progressively emerging. In this review, we compare and integrate findings from rodent and primate studies to create a model of the neural and cellular networks supporting social hierarchies, both from a macro (i.e., circuits) to a micro-scale perspective (microcircuits and synapses). We start by summarizing the literature on the prefrontal cortex and other relevant brain regions to expand the current “prefrontal-centric” view of social hierarchy behaviors. Based on connectivity data we also discuss candidate regions that might inspire further investigation, as well as the caveats and strategies that have been used to further our understanding of the biological substrates underpinning social hierarchy and dominance.
Collapse
Affiliation(s)
- Emanuel Ferreira-Fernandes
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute of Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - João Peça
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- *Correspondence: João Peça
| |
Collapse
|
24
|
Coria-Avila GA, Pfaus JG, Orihuela A, Domínguez-Oliva A, José-Pérez N, Hernández LA, Mota-Rojas D. The Neurobiology of Behavior and Its Applicability for Animal Welfare: A Review. Animals (Basel) 2022; 12:ani12070928. [PMID: 35405916 PMCID: PMC8997080 DOI: 10.3390/ani12070928] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/17/2022] [Accepted: 03/31/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Animal welfare is the result of physical and psychological well-being and is expected to occur if animals are free: (1) from hunger, thirst and malnutrition, (2) from discomfort, (3) from pain, (4) to express normal behavior, and (5) from fear and distress. Nevertheless, well-being is not a constant state but rather the result of certain brain dynamics underlying innate motivated behaviors and learned responses. Thus, by understanding the foundations of the neurobiology of behavior we fathom how emotions and well-being occur in the brain. Herein, we discuss the potential applicability of this approach for animal welfare. First, we provide a general view of the basic responses coordinated by the central nervous system from the processing of internal and external stimuli. Then, we discuss how those stimuli mediate activity in seven neurobiological systems that evoke innate emotional and behavioral responses that directly influence well-being and biological fitness. Finally, we discuss the basic mechanisms of learning and how it affects motivated responses and welfare. Abstract Understanding the foundations of the neurobiology of behavior and well-being can help us better achieve animal welfare. Behavior is the expression of several physiological, endocrine, motor and emotional responses that are coordinated by the central nervous system from the processing of internal and external stimuli. In mammals, seven basic emotional systems have been described that when activated by the right stimuli evoke positive or negative innate responses that evolved to facilitate biological fitness. This review describes the process of how those neurobiological systems can directly influence animal welfare. We also describe examples of the interaction between primary (innate) and secondary (learned) processes that influence behavior.
Collapse
Affiliation(s)
- Genaro A. Coria-Avila
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Avenida Luis Castelazo S/N, Col. Industrial Ánimas, Xalapa 91190, Mexico;
- Correspondence: (G.A.C.-A.); (D.M.-R.)
| | - James G. Pfaus
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Avenida Luis Castelazo S/N, Col. Industrial Ánimas, Xalapa 91190, Mexico;
- Department of Psychology and Life Sciences, Charles University, 182 00 Prague, Czech Republic
- Czech National Institute of Mental Health, 250 67 Klecany, Czech Republic
| | - Agustín Orihuela
- Facultad de Ciencias Agropecuarias, Universidad Autónoma del Estado de Morelos, Cuernavaca 62209, Mexico;
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Unidad Xochimilco, Mexico City 04960, Mexico; (A.D.-O.); (N.J.-P.); (L.A.H.)
| | - Nancy José-Pérez
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Unidad Xochimilco, Mexico City 04960, Mexico; (A.D.-O.); (N.J.-P.); (L.A.H.)
| | - Laura Astrid Hernández
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Unidad Xochimilco, Mexico City 04960, Mexico; (A.D.-O.); (N.J.-P.); (L.A.H.)
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Unidad Xochimilco, Mexico City 04960, Mexico; (A.D.-O.); (N.J.-P.); (L.A.H.)
- Correspondence: (G.A.C.-A.); (D.M.-R.)
| |
Collapse
|
25
|
Ogawa S, Parhar IS. Functions of habenula in reproduction and socio-reproductive behaviours. Front Neuroendocrinol 2022; 64:100964. [PMID: 34793817 DOI: 10.1016/j.yfrne.2021.100964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/11/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022]
Abstract
Habenula is an evolutionarily conserved structure in the brain of vertebrates. Recent reports have drawn attention to the habenula as a processing centre for emotional decision-making and its role in psychiatric disorders. Emotional decision-making process is also known to be closely associated with reproductive conditions. The habenula receives innervations from reproductive centres within the brain and signals from key reproductive neuroendocrine regulators such as gonadal sex steroids, gonadotropin-releasing hormone (GnRH), and kisspeptin. In this review, based on morphological, biochemical, physiological, and pharmacological evidence we discuss an emerging role of the habenula in reproduction. Further, we discuss the modulatory role of reproductive endocrine factors in the habenula and their association with socio-reproductive behaviours such as mating, anxiety and aggression.
Collapse
Affiliation(s)
- Satoshi Ogawa
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia
| | - Ishwar S Parhar
- Brain Research Institute, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, 47500 Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
26
|
Zha X, Xu XH. Neural circuit mechanisms that govern inter-male attack in mice. Cell Mol Life Sci 2021; 78:7289-7307. [PMID: 34687319 PMCID: PMC11072497 DOI: 10.1007/s00018-021-03956-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 10/20/2022]
Abstract
Individuals of many species fight with conspecifics to gain access to or defend critical resources essential for survival and reproduction. Such intraspecific fighting is evolutionarily selected for in a species-, sex-, and environment-dependent manner when the value of resources secured exceeds the cost of fighting. One such example is males fighting for chances to mate with females. Recent advances in new tools open up ways to dissect the detailed neural circuit mechanisms that govern intraspecific, particularly inter-male, aggression in the model organism Mus musculus (house mouse). By targeting and functional manipulating genetically defined populations of neurons and their projections, these studies reveal a core neural circuit that controls the display of reactive male-male attacks in mice, from sensory detection to decision making and action selection. Here, we summarize these critical results. We then describe various modulatory inputs that route into the core circuit to afford state-dependent and top-down modulation of inter-male attacks. While reviewing these exciting developments, we note that how the inter-male attack circuit converges or diverges with neural circuits that mediate other forms of social interactions remain not fully understood. Finally, we emphasize the importance of combining circuit, pharmacological, and genetic analysis when studying the neural control of aggression in the future.
Collapse
Affiliation(s)
- Xi Zha
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiao-Hong Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
27
|
Dimén D, Puska G, Szendi V, Sipos E, Zelena D, Dobolyi Á. Sex-specific parenting and depression evoked by preoptic inhibitory neurons. iScience 2021; 24:103090. [PMID: 34604722 PMCID: PMC8463871 DOI: 10.1016/j.isci.2021.103090] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/20/2021] [Accepted: 09/01/2021] [Indexed: 01/08/2023] Open
Abstract
The role of preoptic GABAergic inhibitory neurons was addressed in parenting, anxiety and depression. Pup exposure and forced swimming resulted in similar c-Fos activation pattern in neurons expressing vesicular GABA transporter in the preoptic area with generally stronger labeling and different distributional pattern in females than in males. Chemogenetic stimulation of preoptic GABAergic cells resulted in elevated maternal motivation and caring behavior in females and mothers but aggression toward pups in males. Behavioral effects were the opposite following inhibition of preoptic GABAergic neurons suggesting their physiological relevance. In addition, increased anxiety-like and depression-like behaviors were found following chemogenetic stimulation of the same neurons in females, whereas previous pup exposure increased only anxiety-like behavior suggesting that not the pups, but overstimulation of the cells can lead to depression-like behavior. A sexually dimorphic projection pattern of preoptic GABAergic neurons was also identified, which could mediate sex-dependent parenting and associated emotional behaviors. Preoptic GABAergic neurons promote maternal behaviors in females mice Activation of preoptic GABAergic neurons induces pup-directed aggression in males Projection pattern of preoptic GABAergic neurons is sexually dimorphic Depression-like behaviors are provoked by stimulation of preoptic GABAergic neurons
Collapse
Affiliation(s)
- Diána Dimén
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Gina Puska
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary.,Department of Ecology, University of Veterinary Medicine Budapest, 1078 Budapest, Hungary
| | - Vivien Szendi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| | - Eszter Sipos
- Department of Behavioral and Stress Studies, Institute of Experimental Medicine, 1080 Budapest, Hungary
| | - Dóra Zelena
- Department of Behavioral and Stress Studies, Institute of Experimental Medicine, 1080 Budapest, Hungary.,Centre for Neuroscience, Szentágothai Research Centre, Institute of Physiology, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Árpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences, Eötvös Loránd Research Network, Eötvös Loránd University, 1117 Budapest, Hungary
| |
Collapse
|
28
|
Smagin DA, Babenko VN, Redina OE, Kovalenko IL, Galyamina AG, Kudryavtseva NN. Reduced Expression of Slc Genes in the VTA and NAcc of Male Mice with Positive Fighting Experience. Genes (Basel) 2021; 12:genes12071099. [PMID: 34356115 PMCID: PMC8306410 DOI: 10.3390/genes12071099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/27/2021] [Accepted: 07/16/2021] [Indexed: 12/28/2022] Open
Abstract
A range of several psychiatric medications targeting the activity of solute carrier (SLC) transporters have proved effective for treatment. Therefore, further research is needed to elucidate the expression profiles of the Slc genes, which may serve as markers of altered brain metabolic processes and neurotransmitter activities in psychoneurological disorders. We studied the Slc differentially expressed genes (DEGs) using transcriptomic profiles in the ventral tegmental area (VTA), nucleus accumbens (NAcc), and prefrontal cortex (PFC) of control and aggressive male mice with psychosis-like behavior induced by repeated experience of aggression accompanied with wins in daily agonistic interactions. The majority of the Slc DEGs were shown to have brain region-specific expression profiles. Most of these genes in the VTA and NAcc (12 of 17 and 25 of 26, respectively) were downregulated, which was not the case in the PFC (6 and 5, up- and downregulated, respectively). In the VTA and NAcc, altered expression was observed for the genes encoding the transporters of neurotransmitters as well as inorganic and organic ions, amino acids, metals, glucose, etc. This indicates an alteration in transport functions for many substrates, which can lead to the downregulation or even disruption of cellular and neurotransmitter processes in the VTA and NAcc, which are attributable to chronic stimulation of the reward systems induced by positive fighting experience. There is not a single Slc DEG common to all three brain regions. Our findings show that in male mice with repeated experience of aggression, altered activity of neurotransmitter systems leads to a restructuring of metabolic and neurotransmitter processes in a way specific for each brain region. We assume that the scoring of Slc DEGs by the largest instances of significant expression co-variation with other genes may outline a candidate for new prognostic drug targets. Thus, we propose that the Slc genes set may be treated as a sensitive genes marker scaffold in brain RNA-Seq studies.
Collapse
Affiliation(s)
- Dmitry A. Smagin
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Vladimir N. Babenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Olga E. Redina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
| | - Irina L. Kovalenko
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Anna G. Galyamina
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
| | - Natalia N. Kudryavtseva
- Neuropathology Modeling Laboratory, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia; (D.A.S.); (V.N.B.); (O.E.R.); (I.L.K.); (A.G.G.)
- Neurogenetics of Social Behavior Sector, The FRC Institute of Cytology and Genetics SB RAS, 630090 Novosibirsk, Russia
- Correspondence:
| |
Collapse
|
29
|
Drosophila Corazonin Neurons as a Hub for Regulating Growth, Stress Responses, Ethanol-Related Behaviors, Copulation Persistence and Sexually Dimorphic Reward Pathways. J Dev Biol 2021; 9:jdb9030026. [PMID: 34287347 PMCID: PMC8293205 DOI: 10.3390/jdb9030026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
The neuronal mechanisms by which complex behaviors are coordinated and timed often involve neuropeptidergic regulation of stress and reward pathways. Recent studies of the neuropeptide Corazonin (Crz), a homolog of the mammalian Gonadotrophin Releasing Hormone (GnRH), have suggested its crucial role in the regulation of growth, internal states and behavioral decision making. We focus this review on Crz neurons with the goal to (1) highlight the diverse roles of Crz neuron function, including mechanisms that may be independent of the Crz peptide, (2) emphasize current gaps in knowledge about Crz neuron functions, and (3) propose exciting ideas of novel research directions involving the use of Crz neurons. We describe the different developmental fates of distinct subsets of Crz neurons, including recent findings elucidating the molecular regulation of apoptosis. Crz regulates systemic growth, food intake, stress responses and homeostasis by interacting with the short Neuropeptide F (sNPF) and the steroid hormone ecdysone. Additionally, activation of Crz neurons is shown to be pleasurable by interacting with the Neuropeptide F (NPF) and regulates reward processes such as ejaculation and ethanol-related behaviors in a sexually dimorphic manner. Crz neurons are proposed to be a motivational switch regulating copulation duration using a CaMKII-dependent mechanism described as the first neuronal interval timer lasting longer than a few seconds. Lastly, we propose ideas to use Crz neuron-induced ejaculation to study the effects of fictive mating and sex addiction in flies, as well as to elucidate dimorphic molecular mechanisms underlying reward behaviors and feeding disorders.
Collapse
|
30
|
Sanfilippo C, Castrogiovanni P, Imbesi R, Lazzarino G, Di Pietro V, Li Volti G, Tibullo D, Barbagallo I, Lazzarino G, Avola R, Musumeci G, Fazio F, Vinciguerra M, Di Rosa M. Sex-dependent monoamine oxidase isoforms expression patterns during human brain ageing. Mech Ageing Dev 2021; 197:111516. [PMID: 34097937 DOI: 10.1016/j.mad.2021.111516] [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: 08/27/2020] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
Human behavior is influenced by both genetic and environmental factors. Monoamine oxidase A (MAOA) is among the most investigated genetic determinants of violent behaviors, while the monoamine oxidase B (MAOB) is explored in Parkinson's disease. We collected twenty-four post-mortem brain tissue datasets of 3871 and 1820 non-demented males and females, respectively, who died from causes not attributable to neurodegenerative diseases. The gene expressions of MAOA and MAOB (MAO genes) were analyzed in these subjects, who were further stratified according to age into eleven groups ranging from late Infancy (5-9 months) to centenarians (>100 years). MAO genes were differently expressed in brains during the entire life span. In particular, maximal and minimal expression levels were found in early life and around the teen years. Females tended to have higher MAO gene levels throughout their lives than those found in age-matched males, even when expressions were separately measured in different brain regions. We demonstrated the existence of age- and sex- related variations in the MAO transcript levels in defined brain regions. More in-depth protein studies are needed to confirm our preliminary results obtained only on messenger RNAs in order to establish the role played by MAO genes in human development.
Collapse
Affiliation(s)
- Cristina Sanfilippo
- IRCCS Centro Neurolesi Bonino Pulejo, Strada Statale 113, C.da Casazza, 98124 Messina, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, School of Medicine, University of Catania, Italy
| | - Rosa Imbesi
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, School of Medicine, University of Catania, Italy
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy
| | - Valentina Di Pietro
- Neurotrauma and Ophthalmology Research Group, Institute of Inflammation and Aging, University of Birmingham, Birmingham B15 2TT, UK
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy
| | - Daniele Tibullo
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy
| | - Ignazio Barbagallo
- Section of Biochemistry, Department of Drug Sciences, University of Catania, 95123 Catania, Italy
| | - Giacomo Lazzarino
- UniCamillus - Saint Camillus International University of Health Sciences, Via di Sant'Alessandro 8, 00131, Rome, Italy
| | - Roberto Avola
- Department of Biomedical and Biotechnological Sciences, Section of Biochemistry, University of Catania, 95123 Catania, Italy
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, School of Medicine, University of Catania, Italy
| | - Francesco Fazio
- University of California San Diego, Department of Psychiatry, Health Science, San Diego La Jolla, CA, USA
| | - Manlio Vinciguerra
- International Clinical Research Center (FNUSA-ICRC), St' Anne University Hospital, Brno, Czech Republic
| | - Michelino Di Rosa
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, School of Medicine, University of Catania, Italy.
| |
Collapse
|
31
|
Zilkha N, Sofer Y, Kashash Y, Kimchi T. The social network: Neural control of sex differences in reproductive behaviors, motivation, and response to social isolation. Curr Opin Neurobiol 2021; 68:137-151. [PMID: 33910083 PMCID: PMC8528716 DOI: 10.1016/j.conb.2021.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/30/2022]
Abstract
Social animal species present a vast repertoire of social interactions when encountering conspecifics. Reproduction-related behaviors, such as mating, parental care, and aggression, are some of the most rewarding types of social interactions and are also the most sexually dimorphic ones. This review focuses on rodent species and summarizes recent advances in neuroscience research that link sexually dimorphic reproductive behaviors to sexual dimorphism in their underlying neuronal circuits. Specifically, we present a few possible mechanisms governing sexually-dimorphic behaviors, by hypothalamic and reward-related brain regions. Sex differences in the neural response to social isolation in adulthood are also discussed, as well as future directions for comparative studies with naturally solitary species.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yizhak Sofer
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Kashash
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
32
|
Nakajo H, Chou MY, Kinoshita M, Appelbaum L, Shimazaki H, Tsuboi T, Okamoto H. Hunger Potentiates the Habenular Winner Pathway for Social Conflict by Orexin-Promoted Biased Alternative Splicing of the AMPA Receptor Gene. Cell Rep 2021; 31:107790. [PMID: 32579920 DOI: 10.1016/j.celrep.2020.107790] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/13/2020] [Accepted: 05/29/2020] [Indexed: 12/31/2022] Open
Abstract
Many animals fight for dominance between conspecifics. Because winners could obtain more resources than losers, fighting outcomes are important for the animal's survival, especially in a situation with insufficient resources, such as hunger. However, it remains unclear whether and how hunger affects fighting outcomes. Herein, we investigate the effects of food deprivation on brain activity and fighting behaviors in zebrafish. We report that starvation induces winning in social conflicts. Before the fights, starved fish show potentiation of the lateral subregion of the dorsal habenula (dHbL)-dorsal/intermediate interpeduncular nucleus (d/iIPN) pathway, which is known to be essential for and potentiated after winning fights. Circuit potentiation is mediated by hypothalamic orexin/hypocretin neuropeptides, which prolong AMPA-type glutamate receptor (AMPAR) activity by increasing the expression of a flip type of alternative splicing variant of the AMPAR subunit. This mechanism may underlie how hungry vertebrates win fights and may be commonly shared across animal phylogeny.
Collapse
Affiliation(s)
- Haruna Nakajo
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan
| | - Ming-Yi Chou
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Masae Kinoshita
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Lior Appelbaum
- The Faculty of Life Sciences and the Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Hideaki Shimazaki
- Graduate School of Informatics, Kyoto University, Kyoto 606-8501, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Center for Brain Science, Saitama 351-0198, Japan; RIKEN CBS-Kao Collaboration Center, Saitama 351-0198, Japan; Department of Life Sciences, Graduate School of Arts and Science, The University of Tokyo, Tokyo 153-8902, Japan.
| |
Collapse
|
33
|
Patel D, Anilkumar S, Chattarji S, de Boer SF, Buwalda B. Repeated victorious and defeat experiences induce similar apical dendritic spine remodeling in CA1 hippocampus of rats. Behav Brain Res 2021; 406:113243. [PMID: 33727049 DOI: 10.1016/j.bbr.2021.113243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 02/24/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022]
Abstract
In this study, apical dendritic spine density of neurons in hippocampal, amygdalar and prefrontal cortical areas was compared in rats that were repeatedly winning or losing social conflicts. Territorial male wild-type Groningen (WTG) rats were allowed multiple daily attacks (>20 times) on intruder males in the resident-intruder paradigm. Frequent winning experiences are known to facilitate uncontrolled aggressive behavior reflected in aggressive attacks on anesthetized males which was also observed in the winners in this study. Both winners and losers were socially housed during the experiments; winners with females to stimulate territorial behavior, and losers with two other losing male rats. Twenty-four hours after the last social encounter, brains from experienced residential winners and repeatedly defeated intruder rats were collected and neuronal morphology in selected brain regions was studied via Golgi-Cox staining. Results indicate that spine density in the apical dendrites of the hippocampal CA1 reduced similarly in both winners and losers. In addition, winners showed increased spine densities at the proximal segments (20-30 μm) of the basolateral amygdala neurons and losers tended to show a decreased spine density at the more proximal segments of the infralimbic region of prefrontal cortex neurons. No effect of winning and losing was observed in the medial amygdala. The atrophic effect of repeated defeats in hippocampal and prefrontal regions was anticipated despite the fact that social housing of the repeatedly losing intruder males may have played a protective role. The reduction of hippocampal spine density in the winners seems surprising but supports previous findings in hierarchical dominant males in rat colonies. The dominants showed even greater shrinkage of the apical dendritic arbors of hippocampal CA3 pyramidal neurons compared to the stressed subordinates.
Collapse
Affiliation(s)
- Deepika Patel
- National Centre for Biological Sciences, Bangalore, 560065, India; Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Shobha Anilkumar
- National Centre for Biological Sciences, Bangalore, 560065, India; Manipal University, Manipal, India
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Bangalore, 560065, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India; Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH89XD, UK
| | - Sietse F de Boer
- Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Bauke Buwalda
- Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
34
|
Coley AA, Padilla-Coreano N, Patel R, Tye KM. Valence processing in the PFC: Reconciling circuit-level and systems-level views. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 158:171-212. [PMID: 33785145 DOI: 10.1016/bs.irn.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
An essential component in animal behavior is the ability to process emotion and dissociate among positive and negative valence in response to a rewarding or aversive stimulus. The medial prefrontal cortex (mPFC)-responsible for higher order executive functions that include cognition, learning, and working memory; and is also involved in sociability-plays a major role in emotional processing and control. Although the amygdala is widely regarded as the "emotional hub," the mPFC encodes for context-specific salience and elicits top-down control over limbic circuitry. The mPFC can then conduct behavioral responses, via cortico-striatal and cortico-brainstem pathways, that correspond to emotional stimuli. Evidence shows that abnormalities within the mPFC lead to sociability deficits, working memory impairments, and drug-seeking behavior that include addiction and compulsive disorders; as well as conditions such as anhedonia. Recent studies investigate the effects of aberrant salience processing on cortical circuitry and neuronal populations associated with these behaviors. In this chapter, we discuss mPFC valence processing, neuroanatomical connections, and physiological substrates involved in mPFC-associated behavior. We review neurocomputational and theoretical models such as "mixed selectivity," that describe cognitive control, attentiveness, and motivational drives. Using this knowledge, we describe the effects of valence imbalances and its influence on mPFC neural pathways that contribute to deficits in social cognition, while understanding the effects in addiction/compulsive behaviors and anhedonia.
Collapse
Affiliation(s)
- Austin A Coley
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | | | - Reesha Patel
- Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Kay M Tye
- Salk Institute for Biological Studies, La Jolla, CA, United States.
| |
Collapse
|
35
|
Reichmann F, Rimmer N, Tilley CA, Dalla Vecchia E, Pinion J, Al Oustah A, Carreño Gutiérrez H, Young AMJ, McDearmid JR, Winter MJ, Norton WHJ. The zebrafish histamine H3 receptor modulates aggression, neural activity and forebrain functional connectivity. Acta Physiol (Oxf) 2020; 230:e13543. [PMID: 32743878 DOI: 10.1111/apha.13543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 01/17/2023]
Abstract
AIM Aggression is a behavioural trait characterized by the intention to harm others for offensive or defensive purposes. Neurotransmitters such as serotonin and dopamine are important mediators of aggression. However, the physiological role of the histaminergic system during this behaviour is currently unclear. Here, we aimed to better understand histaminergic signalling during aggression by characterizing the involvement of the histamine H3 receptor (Hrh3). METHODS We have generated a novel zebrafish Hrh3 null mutant line using CRISPR-Cas9 genome engineering and investigated behavioural changes and alterations to neural activity using whole brain Ca2+ imaging in zebrafish larvae and ribosomal protein S6 (rpS6) immunohistochemistry in adults. RESULTS We show that genetic inactivation of the histamine H3 receptor (Hrh3) reduces aggression in zebrafish, an effect that can be reproduced by pharmacological inhibition. In addition, hrh3-/- zebrafish show behavioural impairments consistent with heightened anxiety. Larval in vivo whole brain Ca2+ imaging reveals higher neuronal activity in the forebrain of mutants, but lower activity in specific hindbrain areas and changes in measures of functional connectivity between subregions. Adult hrh3-/- zebrafish display brain region-specific neural activity changes in response to aggression of both key regions of the social decision-making network, and the areas containing histaminergic neurons in the zebrafish brain. CONCLUSION These results highlight the importance of zebrafish Hrh3 signalling for aggression and anxiety and uncover the brain areas involved. Targeting this receptor might be a potential novel therapeutic route for human conditions characterized by heightened aggression.
Collapse
Affiliation(s)
- Florian Reichmann
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
- Division of Pharmacology, Otto Loewi Research Centre for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Neal Rimmer
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Ceinwen A Tilley
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Elisa Dalla Vecchia
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Joseph Pinion
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Amir Al Oustah
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Hector Carreño Gutiérrez
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Andrew M J Young
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Jonathan R McDearmid
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
| | - Matthew J Winter
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, College of Life Sciences, University of Leicester, Leicester, UK
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| |
Collapse
|
36
|
The drawbacks of Information and Communication Technologies: Interplay and psychopathological risk of nomophobia and cyber-bullying, results from the bullying and youth mental health Naples study (BYMHNS). COMPUTERS IN HUMAN BEHAVIOR 2020. [DOI: 10.1016/j.chb.2020.106496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
37
|
Anilkumar S, Patel D, de Boer SF, Chattarji S, Buwalda B. Decreased dendritic spine density in posterodorsal medial amygdala neurons of proactive coping rats. Behav Brain Res 2020; 397:112940. [PMID: 33126115 DOI: 10.1016/j.bbr.2020.112940] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 09/02/2020] [Accepted: 09/22/2020] [Indexed: 01/30/2023]
Abstract
There are large individual differences in the way animals, including humans, behaviorally and physiologically cope with environmental challenges and opportunities. Rodents with either a proactive or reactive coping style not only differ in their capacity to adapt successfully to environmental conditions, but also have a differential susceptibility to develop stress-related (psycho)pathologies when coping fails. In this study, we explored if there are structural neuronal differences in spine density in brain regions important for the regulation of stress coping styles. For this, the individual coping styles of wild-type Groningen (WTG) rats were determined using their level of offensive aggressiveness assessed in the resident-intruder paradigm. Subsequently, brains from proactive (high-aggressive) and reactive (low-aggressive) rats were Golgi-cox stained for spine quantification. The results reveal that dendritic spine densities in the dorsal hippocampal CA1 region and basolateral amygdala are similar in rats with proactive and reactive coping styles. Interestingly, however, dendritic spine density in the medial amygdala (MeA) is strikingly reduced in the proactive coping rats. This brain region is reported to be strongly involved in rivalry aggression which is the criterion by which the coping styles in our study are dissociated. The possibility that structural differences in spine density in the MeA are involved in other behavioral traits of distinct coping styles needs further investigation.
Collapse
Affiliation(s)
- Shobha Anilkumar
- National Centre for Biological Sciences, Bangalore, 560065, India; Manipal University, Manipal, India
| | - Deepika Patel
- National Centre for Biological Sciences, Bangalore, 560065, India; Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Sietse F de Boer
- Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Bangalore, 560065, India; Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, 560065, India; Centre for Integrative Physiology, Deanery of Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH89XD, UK
| | - Bauke Buwalda
- Dept. of Behavioral Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
38
|
Zhang L, Hu X, Lu L, Li B, Hu X, Bu X, Li H, Tang S, Gao Y, Yang Y, Sweeney JA, Gong Q, Huang X. Anatomic alterations across amygdala subnuclei in medication-free patients with obsessive-compulsive disorder. J Psychiatry Neurosci 2020; 45:334-343. [PMID: 32293840 PMCID: PMC7850150 DOI: 10.1503/jpn.190114] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The amygdala has been implicated in obsessive-compulsive disorder (OCD), a common, disabling illness. However, the regional distribution of anatomic alterations in this structure and their association with the symptoms of OCD remains to be established. METHODS We collected high-resolution 3D T1-weighted images from 81 untreated patients with OCD and no lifetime history of comorbid psychotic, affective or anxiety disorders, and from 95 age- and sex-matched healthy controls. We extracted the volume of the central nucleus of the amygdala (CeA) and the basolateral complex of the amygdala (BLA) and compared them across groups using FreeSurfer 6.0. In exploratory analyses, we evaluated other subnuclei, including the cortical medial nuclei, the anterior amygdaloid area, and the corticoamygdaloid transition area. RESULTS Patients with OCD had reduced amygdala volume bilaterally compared with healthy controls (left, p = 0.034; right, p = 0.002). Volume reductions were greater in the CeA (left: -11.9%, p = 0.002; right: -13.3%, p < 0.001) than in the BLA (left lateral nucleus: -3.3%, p = 0.029; right lateral nucleus: -3.9%, p = 0.018; right basal nucleus: -4.1%, p = 0.017; left accessory basal nucleus: -6.5%, p = 0.001; right accessory basal nucleus: -9.3%, p < 0.001). Volume reductions in the CeA were associated with illness duration. Exploratory analysis revealed smaller medial (left: -15.4%, p < 0.001, η2 = 0.101) and cortical (left: -9.1%, p = 0.001, η2 = 0.058; right: -15.4%, p < 0.001, η2 = 0.175) nuclei in patients with OCD compared with healthy controls. LIMITATIONS Although the strict exclusion criteria used in the study helped us to identify OCD-specific alterations, they may have limited generalizability to the broader OCD population. CONCLUSION Our results provide a comprehensive anatomic profile of alterations in the amygdala subnuclei in untreated patients with OCD and highlight a distinctive pattern of volume reductions across subnuclei in OCD. Based on the functional properties of the amygdala subnuclei established from preclinical research, CeA impairment may contribute to behavioural inflexibility, and BLA disruption may be responsible for altered fear conditioning and the affective components of OCD.
Collapse
Affiliation(s)
- Lianqing Zhang
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Xinyu Hu
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Lu Lu
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Bin Li
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Xiaoxiao Hu
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Xuan Bu
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Hailong Li
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Shi Tang
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Yingxue Gao
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Yanchun Yang
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - John A Sweeney
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Qiyong Gong
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| | - Xiaoqi Huang
- From the Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Sweeney, Gong, Huang); the Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, PR China (Li, Yang); the Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, USA (Sweeney); and the Psychoradiology Research Unit of the Chinese Academy of Medical Sciences (2018RU011), West China Hospital of Sichuan University, Chengdu, Sichuan, China (Zhang, Xinyu Hu, Lu, Xiaoxiao Hu, Bu, Li, Tang, Gao, Gong, Huang)
| |
Collapse
|
39
|
Helmy M, Zhang J, Wang H. Neurobiology and Neural Circuits of Aggression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1284:9-22. [DOI: 10.1007/978-981-15-7086-5_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Abstract
A recent study has shown that local inhibitory GAD2-positive neurons regulate the activity of lateral habenula neurons, thereby governing aggressive behavior in male mice.
Collapse
Affiliation(s)
- Jack F Webster
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Christian Wozny
- Strathclyde Institute for Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
41
|
Pulver A, Kiive E, Kanarik M, Harro J. Association of orexin/hypocretin receptor gene (HCRTR1) with reward sensitivity, and interaction with gender. Brain Res 2020; 1746:147013. [PMID: 32652147 DOI: 10.1016/j.brainres.2020.147013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
Orexins/hypocretins maintain wakefulness, increase appetite and participate in the coordination of stress response. We have recently provided evidence on the role of orexins in aggression, showing the association of the HCRTR1 genotype. (rs2271933 G > A; leading to amino acid substitution Ile408Val) with aggressiveness or breach of law in four independent cohorts. Aggressive behaviour can be reward driven and hence we have examined the association of HCRTR1 rs2271933 genotype with different aspects of reward sensitivity in the birth cohort representative Estonian Children Personality Behaviour and Health Study. HCRTR1 genotype was associated with reward sensitivity in a gender dependent manner. Male HCRTR1 A/A homozygotes had higher Openness to Rewards and the overall reward sensitivity score while, in contrast, female A/A homozygotes scored lower than G-allele carriers in Openness to Rewards. In the total sample, aggressiveness correlated positively with reward sensitivity, but this was on account of Insatiability by Reward. In contrast, the HCRTR1 A/A homozygotes had a positive association of aggressiveness and Openness to Rewards. Experience of stressful life events had a small but significant increasing effect on both aspects of reward sensitivity, and correlated in an anomalous way with reward sensitivity in the HCRTR1 A/A homozygotes. Conclusively, the higher aggressiveness of HCRTR1 A/A homozygotes appears based on a qualitative difference in sensitivity to rewards, in the form that suggests their lower ability to prevent responses to challenges being converted into overt aggression.
Collapse
Affiliation(s)
- Aleksander Pulver
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, Astra Building, 10120 Tallinn, Estonia
| | - Evelyn Kiive
- Division of Special Education, Department of Education, University of Tartu, Näituse 2, 50409 Tartu, Estonia
| | - Margus Kanarik
- Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia
| | - Jaanus Harro
- School of Natural Sciences and Health, Tallinn University, Narva Road 29, Astra Building, 10120 Tallinn, Estonia; Division of Neuropsychopharmacology, Department of Psychology, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia.
| |
Collapse
|
42
|
Lewis AS, Picciotto MR. Regulation of aggressive behaviors by nicotinic acetylcholine receptors: Animal models, human genetics, and clinical studies. Neuropharmacology 2020; 167:107929. [PMID: 32058178 PMCID: PMC7080580 DOI: 10.1016/j.neuropharm.2019.107929] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/18/2019] [Accepted: 12/25/2019] [Indexed: 12/12/2022]
Abstract
Neuropsychiatric disorders are frequently complicated by aggressive behaviors. For some individuals, existing behavioral and psychopharmacological treatments are ineffective or confer significant side effects, necessitating development of new ways to treat patients with severe aggression. Nicotinic acetylcholine receptors (nAChRs) are a large and diverse family of ligand-gated ion channels expressed throughout the brain that influence behaviors highly relevant for neuropsychiatric disorders, including attention, mood, and impulsivity. Nicotine and other drugs targeting nAChRs can reduce aggression in animal models of offensive, defensive, and predatory aggression, as well as in human laboratory studies. Human genetic studies have suggested a relationship between the CHRNA7 gene encoding the alpha-7 nAChR and aggressive behavior, although these effects are heterogeneous and strongly influenced by genetic background and environment. Here we review animal, human genetic, and clinical studies supporting a consistent role of nicotine and nAChR signaling in modulation of aggressive behaviors. We integrate findings from recent studies of aggression neuroscience, discuss the circuitry that may be involved in these effects of nAChRs, and identify multiple key questions that must be answered prior to safe and effective translation for human patients. This article is part of the special issue on 'Contemporary Advances in Nicotine Neuropharmacology'.
Collapse
Affiliation(s)
- Alan S Lewis
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Cognitive Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| | | |
Collapse
|
43
|
Eshel N, Leibenluft E. New Frontiers in Irritability Research-From Cradle to Grave and Bench to Bedside. JAMA Psychiatry 2020; 77:227-228. [PMID: 31799997 DOI: 10.1001/jamapsychiatry.2019.3686] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Neir Eshel
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, California
| | - Ellen Leibenluft
- Section on Mood Dysregulation and Neuroscience, Intramural Research Program, National Institute of Mental Health, Bethesda, Maryland
| |
Collapse
|
44
|
Neural circuits for coping with social defeat. Curr Opin Neurobiol 2019; 60:99-107. [PMID: 31837481 DOI: 10.1016/j.conb.2019.11.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 12/27/2022]
Abstract
When resources, such as food, territory, and potential mates are limited, competition among animals of the same species is inevitable. Over bouts of agonistic interactions, winners and losers are determined. Losing is a traumatic experience, both physically and psychologically. Losers not only need to deploy a set of species-specific defensive behaviors to minimize the physical damage during defeat, but also adjust their behavior towards the winners to avoid future fights in which they are likely disadvantaged. The expression of defensive behaviors and the fast and long-lasting changes in behaviors accompanying defeat must be supported by a complex neural circuit. This review summarizes the brain regions that have been implicated in coping with social defeat, one centered on basolateral amygdala and the other on ventromedial hypothalamus. Gaps in our knowledge and hypotheses that may help guide future experiments are also discussed.
Collapse
|
45
|
Nucleus Accumbens Dopamine Receptor 1 Expressing Neurons Are Instrumental in Appetitive Aggression. J Neurosci 2019; 39:6610-6612. [PMID: 31434715 DOI: 10.1523/jneurosci.0772-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/15/2019] [Accepted: 06/21/2019] [Indexed: 11/21/2022] Open
|
46
|
Kim EC, Patel J, Zhang J, Soh H, Rhodes JS, Tzingounis AV, Chung HJ. Heterozygous loss of epilepsy gene KCNQ2 alters social, repetitive and exploratory behaviors. GENES BRAIN AND BEHAVIOR 2019; 19:e12599. [PMID: 31283873 PMCID: PMC7050516 DOI: 10.1111/gbb.12599] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/28/2019] [Accepted: 07/06/2019] [Indexed: 12/28/2022]
Abstract
KCNQ/Kv7 channels conduct voltage‐dependent outward potassium currents that potently decrease neuronal excitability. Heterozygous inherited mutations in their principle subunits Kv7.2/KCNQ2 and Kv7.3/KCNQ3 cause benign familial neonatal epilepsy whereas patients with de novo heterozygous Kv7.2 mutations are associated with early‐onset epileptic encephalopathy and neurodevelopmental disorders characterized by intellectual disability, developmental delay and autism. However, the role of Kv7.2‐containing Kv7 channels in behaviors especially autism‐associated behaviors has not been described. Because pathogenic Kv7.2 mutations in patients are typically heterozygous loss‐of‐function mutations, we investigated the contributions of Kv7.2 to exploratory, social, repetitive and compulsive‐like behaviors by behavioral phenotyping of both male and female KCNQ2+/− mice that were heterozygous null for the KCNQ2 gene. Compared with their wild‐type littermates, male and female KCNQ2+/− mice displayed increased locomotor activity in their home cage during the light phase but not the dark phase and showed no difference in motor coordination, suggesting hyperactivity during the inactive light phase. In the dark phase, KCNQ2+/− group showed enhanced exploratory behaviors, and repetitive grooming but decreased sociability with sex differences in the degree of these behaviors. While male KCNQ2+/− mice displayed enhanced compulsive‐like behavior and social dominance, female KCNQ2+/− mice did not. In addition to elevated seizure susceptibility, our findings together indicate that heterozygous loss of Kv7.2 induces behavioral abnormalities including autism‐associated behaviors such as reduced sociability and enhanced repetitive behaviors. Therefore, our study is the first to provide a tangible link between loss‐of‐function Kv7.2 mutations and the behavioral comorbidities of Kv7.2‐associated epilepsy.
Collapse
Affiliation(s)
- Eung Chang Kim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jaimin Patel
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jiaren Zhang
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Heun Soh
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut
| | - Justin S Rhodes
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Psychology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | - Hee Jung Chung
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
47
|
Kompier NF, Keysers C, Gazzola V, Lucassen PJ, Krugers HJ. Early Life Adversity and Adult Social Behavior: Focus on Arginine Vasopressin and Oxytocin as Potential Mediators. Front Behav Neurosci 2019; 13:143. [PMID: 31404254 PMCID: PMC6676334 DOI: 10.3389/fnbeh.2019.00143] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/11/2019] [Indexed: 01/23/2023] Open
Abstract
Exposure to stress during the early postnatal period (i.e., early life stress, ES) can impact brain physiology and modify individual variability in adult social behavior. Arginine vasopressin (AVP) and oxytocin (OXT) are two centrally released neuropeptides that are involved in shaping essential social behaviors, like aggression, social recognition, and social motivation. AVP and OXT modulate activity in brain regions important for the establishment of social behavior, and may be particularly sensitive to ES. In this review, we discuss whether ES alters the characteristics of the AVP- and OXT- systems in rodents, and whether these changes are associated with later alterations in aggression, social recognition, and social motivation. We have integrated causal studies indicating that (1) ES affects AVP/OXT, and (2) that changing AVP/OXT in affected regions alters social behavior. Although there is encouraging evidence that ES causes AVP- and OXT-system changes, and that these may mediate social behavior, a comprehensive understanding of the exact nature of AVP- and OXT changes and whether they are causal in establishing these behavioral disturbances needs further investigation. As there are indications that ES alters AVP- and OXT characteristics in humans as well, and that these may interact with adult predisposition to psychopathology with social dysfunction, future rodent studies may lay ground for a better understanding of such changes in humans. Ultimately, this may assist in developing therapeutic strategies to target ES effects on social behavior.
Collapse
Affiliation(s)
- Nine F. Kompier
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
- Social Brain Lab, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Christian Keysers
- Social Brain Lab, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Valeria Gazzola
- Social Brain Lab, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- Department of Psychology, University of Amsterdam, Amsterdam, Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Harmen J. Krugers
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
48
|
Locci A, Pinna G. Social isolation as a promising animal model of PTSD comorbid suicide: neurosteroids and cannabinoids as possible treatment options. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:243-259. [PMID: 30586627 DOI: 10.1016/j.pnpbp.2018.12.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/18/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric condition characterized by drastic alterations in mood, emotions, social abilities and cognition. Notably, one aspect of PTSD, particularly in veterans, is its comorbidity with suicide. Elevated aggressiveness predicts high-risk to suicide in humans and despite the difficulty in reproducing a complex human suicidal behavior in rodents, aggressive behavior is a well reproducible behavioral trait of suicide. PTSD animal models are based on a peculiar phenotype, including exaggerated fear memory and impaired fear extinction associated with neurochemical dysregulations in the brain circuitry regulating emotion. The endocannabinoid and the neurosteroid systems regulate emotions and stress responses, and recent evidence shows these two systems are interrelated and critically compromised in neuropsychiatric disorders. For instance, levels of the neurosteroid, allopregnanolone, as well as those of the endocannabinoids, anandamide and its congener, palmitoylethanolamide are decreased in PTSD. Similarly, the endocannabinoid system and neurosteroid biosynthesis are altered in suicidal individuals. Selective serotonin reuptake inhibitors (SSRIs), the only FDA-approved treatments for PTSD, fail to help half of the treatment-seeking patients. This highlights the need for developing biomarker-based efficient therapies. One promising alternative to SSRIs points to stimulation of allopregnanolone biosynthesis as a treatment and a valid end-point to predict treatment response in PTSD patients. This review highlights running findings on the role of the endocannabinoid and neurosteroid systems in PTSD and suicidal behavior both in a preclinical and clinical perspective. A specific focus is given to predictive PTSD/suicide animal models. Ultimately, we discuss the idea that disruption of neurosteroid and endocannabinoid biosynthesis may offer a novel promising biomarker axis to develop new treatments for PTSD and, perhaps, suicidal behavior.
Collapse
Affiliation(s)
- Andrea Locci
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 1601 W. Taylor St., Chicago, IL 60612, USA
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois at Chicago, 1601 W. Taylor St., Chicago, IL 60612, USA.
| |
Collapse
|
49
|
Animal Models of (or for) Aggression Reward, Addiction, and Relapse: Behavior and Circuits. J Neurosci 2019; 39:3996-4008. [PMID: 30833504 DOI: 10.1523/jneurosci.0151-19.2019] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/18/2019] [Accepted: 02/25/2019] [Indexed: 02/06/2023] Open
Abstract
Inappropriate and pathological aggression plays a leading role in the suffering and death of millions of people, and further places an untenable strain on the caregivers and families of those afflicted. In some cases, such as addictive drugs, aggression can be highly rewarding (appetitive) and continually pursued despite short- and long-term negative consequences. Similarly, recidivism (relapse) rates for repeat violent offenders are as high as relapse rates for drug addicts. Appetitive aggression and relapse to aggression seeking can be modeled in mice studies using conditioned place preference and self-administration procedures followed by a period of abstinence and subsequent tests for relapse to aggression preference and aggression seeking. These procedures allow for the study of the mechanisms that control the appetitive versus the consummatory (attack) phases of aggressive behavior. In this review, we first discuss the behavioral procedures developed to probe appetitive aggression in mouse models, spanning from Pavlovian to operant tasks, and we also describe the recently proposed phenomenon of "aggression addiction." Next, we discuss the pharmacological and circuit mechanisms of aggression conditioned place preference and aggression self-administration, seeking, and relapse, highlighting mechanistic congruence and divergence between appetitive and consummatory phases of aggression. We conclude by discussing clinical implications of the studies reviewed.
Collapse
|
50
|
Nucleus Accumbens Drd1-Expressing Neurons Control Aggression Self-Administration and Aggression Seeking in Mice. J Neurosci 2019; 39:2482-2496. [PMID: 30655356 DOI: 10.1523/jneurosci.2409-18.2019] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/21/2018] [Accepted: 01/07/2019] [Indexed: 01/01/2023] Open
Abstract
We recently developed a mouse model of appetitive operant aggression and reported that adult male outbred CD-1 mice lever-press for the opportunity to attack subordinate male mice and relapse to aggression seeking during abstinence. Here we studied the role of nucleus accumbens (NAc) dopamine receptor (Drd)1- and Drd2-expressing neurons in aggression self-administration and aggression seeking. We trained CD-1 mice to self-administer intruders (9 d, 12 trials/d) and tested them for aggression self-administration and aggression seeking on abstinence Day 1. We used immunohistochemistry and in situ hybridization to measure the neuronal activity marker Fos in the NAc, and cell-type-specific colocalization of Fos with Drd1- and Drd2-expressing neurons. To test the causal role of Drd1- and Drd2-expressing neurons, we validated a transgenic hybrid breeding strategy crossing inbred Drd1-Cre and Drd2-Cre transgenic mice with outbred CD-1 mice and used cell-type-specific Cre-DREADD (hM4Di) to inhibit NAc Drd1- and Drd2-expressing neuron activity. We found that aggression self-administration and aggression seeking induced higher Fos expression in NAc shell than in core, that Fos colocalized with Drd1 and Drd2 in both subregions, and that chemogenetic inhibition of Drd1-, but not Drd2-, expressing neurons decreased aggression self-administration and aggression seeking. Results indicate a cell-type-specific role of Drd1-expressing neurons that is critical for both aggression self-administration and aggression seeking. Our study also validates a simple breeding strategy between outbred CD-1 mice and inbred C57-based Cre lines that can be used to study cell-type and circuit mechanisms of aggression reward and relapse.SIGNIFICANCE STATEMENT Aggression is often comorbid with neuropsychiatric diseases, including drug addiction. One form, appetitive aggression, exhibits symptomatology that mimics that of drug addiction and is hypothesized to be due to dysregulation of addiction-related reward circuits. However, our mechanistic understanding of the circuitry modulating appetitive operant aggression is limited. Here we used a novel mouse model of aggression self-administration and relapse, in combination with immunohistochemistry, in situ hybridization, and chemogenetic manipulations to examine how cell types in the nucleus accumbens are recruited for, and control, operant aggression self-administration and aggression seeking on abstinence Day 1. We found that one population, dopamine receptor 1-expressing neurons, act as a critical modulator of operant aggression reward and aggression seeking.
Collapse
|