1
|
Zafar S, Williams C, Joo J, Himes BE, Schneider JS. Developmental lead exposure and aggression in male rats: Influences of maternal care and environmental enrichment. Toxicol Rep 2025; 14:101937. [PMID: 39989979 PMCID: PMC11847132 DOI: 10.1016/j.toxrep.2025.101937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/18/2025] [Accepted: 01/29/2025] [Indexed: 02/25/2025] Open
Abstract
Developmental lead (Pb) exposure results in a variety of cognitive deficits and behavioral issues including increased antisocial behavior and aggression. This study investigated the effect of developmental Pb exposure on aggression and violent behavior in male rats and the potential modulatory influences of quality of maternal care and enriched/non-enriched housing conditions. Long-Evans male rats with or without Pb exposure (perinatal or early postnatal) from low or high maternal care mothers (based on amounts of licking/grooming and arched-back nursing) were randomly assigned to live in enriched or non-enriched environments at weaning. At postnatal day 120-190, offensive aggression was assessed using a resident intruder test. Clinch attack (CAK) frequency and latency, and occurrence of biting events were observed to determine violent behavior. Both perinatal and postnatal Pb-exposed rats were significantly more aggressive and showed more violent behavior, compared to non-Pb-exposed animals, regardless of level of maternal care and environmental enrichment. High maternal care significantly lowered the proportion of animals with short CAK latencies and enriched housing significantly lowered the occurrence of biting events. These results suggest that high maternal care and enriched housing may potentially modify expression of violent aggressive behavior in rats with early life Pb exposure.
Collapse
Affiliation(s)
- Shamaila Zafar
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Courtney Williams
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Jaehyun Joo
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Blanca E. Himes
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jay S. Schneider
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, United States
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
2
|
Mikulovic S, Lenschow C. Neural control of sex differences in affiliative and prosocial behaviors. Neurosci Biobehav Rev 2025; 171:106039. [PMID: 39914700 DOI: 10.1016/j.neubiorev.2025.106039] [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: 06/12/2024] [Revised: 01/28/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Social interactions are vital for various taxa and species. Prosocial and affiliative dynamics within a group and between individuals are not only pleasurable and rewarding, but also appear to actively contribute to well-being, cognitive performance, and disease prevention. Moreover, disturbances in acting or being prosocial can represent a major burden for an individual and their affective partners. These disruptions are evident across a spectrum of neuropsychiatric conditions, including depression and autism spectrum disorders. Importantly, interactive patterns of prosocial and affiliative behavior can vary with sex. The fact that genders are differentially affected by neuropsychiatric disorders associated with social impairment underscores the high importance of this research in uncovering the underlying neural correlates and mechanisms. This review focuses on elucidating sex-related differences in prosocial and affiliative behaviors and their potential association with sexually different neural correlates. Specifically, we aim to shed light on the complex interplay between sex, behavior, and neurobiology in affiliative and prosocial interaction patterns.
Collapse
Affiliation(s)
- Sanja Mikulovic
- Leibniz Institute for Neurobiology, Brennecke Straße, Magdeburg, Germany.
| | - Constanze Lenschow
- Otto-von-Guericke University Magdeburg, Institute of Biology (House 91), Leipziger Straße 44, Magdeburg 39120, Germany.
| |
Collapse
|
3
|
Dai B, Zheng B, Dai X, Cui X, Yin L, Cai J, Zhuo Y, Tritsch NX, Zweifel LS, Li Y, Lin D. Experience-dependent dopamine modulation of male aggression. Nature 2025; 639:430-437. [PMID: 39843745 DOI: 10.1038/s41586-024-08459-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 11/27/2024] [Indexed: 01/24/2025]
Abstract
Numerous studies support the role of dopamine in modulating aggression1,2, but the exact neural mechanisms remain elusive. Here we show that dopaminergic cells in the ventral tegmental area (VTA) can bidirectionally modulate aggression in male mice in an experience-dependent manner. Although VTA dopaminergic cells strongly influence aggression in novice aggressors, they become ineffective in expert aggressors. Furthermore, eliminating dopamine synthesis in the VTA prevents the emergence of aggression in naive mice but leaves aggression intact in expert aggressors. VTA dopamine modulates aggression through the dorsal lateral septum (dLS), a region known for aggression control. Dopamine enables the flow of information from the hippocampus to the dLS by weakening local inhibition in novice aggressors. In expert aggressors, dLS local inhibition naturally weakens, and the ability of dopamine to modulate dLS cells diminishes. Overall, these results reveal a sophisticated role of dopamine in the rise of aggression in adult male mice.
Collapse
Affiliation(s)
- Bing Dai
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| | - Bingqin Zheng
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Xiuzhi Dai
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Xiaoyang Cui
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Luping Yin
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Jing Cai
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yizhou Zhuo
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Department of Psychiatry, Douglas Hospital Research Centre, McGill University, Montreal, Quebec, Canada
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, USA
- Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Dayu Lin
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Chen H, Ferguson CJ, Mitchell DC, Risch I, Titus A, Paulo JA, Hwang A, Beck LK, Lin TH, Gu W, Song SK, Yuede CM, Yano H, Griffith OL, Griffith M, Gygi SP, Bonni A, Kim AH. The Hao-Fountain syndrome protein USP7 regulates neuronal connectivity in the brain via a novel p53-independent ubiquitin signaling pathway. Cell Rep 2025; 44:115231. [PMID: 39862434 PMCID: PMC11922642 DOI: 10.1016/j.celrep.2025.115231] [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: 01/06/2024] [Revised: 11/14/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Mutation or deletion of the deubiquitinase USP7 causes Hao-Fountain syndrome (HAFOUS), which is characterized by speech delay, intellectual disability, and aggressive behavior and highlights important unknown roles of USP7 in the nervous system. Here, we conditionally delete USP7 in glutamatergic neurons in the mouse forebrain, triggering disease-relevant phenotypes, including sensorimotor deficits, impaired cognition, and aggressive behavior. Although USP7 deletion induces p53-dependent neuronal apoptosis, most behavioral abnormalities in USP7 conditional knockout mice persist following p53 loss. Strikingly, USP7 deletion perturbs the synaptic proteome and dendritic spinogenesis independent of p53. Integrated proteomics and biochemical analyses identify the RNA splicing factor Ppil4 as a key substrate of USP7. Ppil4 knockdown phenocopies the effect of USP7 loss on dendritic spines. Accordingly, USP7 loss disrupts splicing of synaptic genes. These findings reveal that USP7-Ppil4 signaling regulates neuronal connectivity in the developing brain with implications for our understanding of HAFOUS pathogenesis and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cole J Ferguson
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Dylan C Mitchell
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Isabel Risch
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Amanda Titus
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Hwang
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Loren K Beck
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tsen-Hsuan Lin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Gu
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Sheng-Kwei Song
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carla M Yuede
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Hiroko Yano
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Obi L Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Malachi Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Roche Pharma Research and Early Development, Neuroscience and Rare Disease Discovery and Translational Area, Roche Innovation Center, 4070 Basel, Switzerland.
| | - Albert H Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
5
|
Su C, Zhang L, Pan Y, Jiao J, Luo P, Chang X, Zhang H, Si X, Chen W, Huang Y. Enhancing aggression in Henan gamecocks via augmentation of serotonergic-dopaminergic signaling and attenuation of neuroimmune response. Poult Sci 2024; 103:104055. [PMID: 39190992 PMCID: PMC11395772 DOI: 10.1016/j.psj.2024.104055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/18/2024] [Accepted: 06/27/2024] [Indexed: 08/29/2024] Open
Abstract
Animal aggression is one of the most conserved behaviors. Excessive and inappropriate aggression was a serious social concern across species. After long-term selection under strict stress conditions, Henan gamecock serves as a good model for studying aggressive behavior. In this research, we constructed a Henan game chicken backcross population containing 25% Rhode Island Red (RIR), and conducted brain transcriptomics and serum metabolomics analyses on Henan gamecock (HGR) through its comparison with its female encounters (HGH) and the male backcross birds (BGR). The study revealed that seven differential metabolites in serum and 172 differentially expressed genes in the brain were commonly shared in both HGR vs. HGH and HGR vs. BGR comparisons. They exhibited the same patterns of modulation in Henan gamecocks, following either HGH < HGR > BGR or HGH > HGR < BGR style. Therein, some neurological genes involving in serotonergic and dopaminergic signaling were upregulated, while the levels of many genes related with neuro-immune function were decreased in Henan gamecock. In addition, many unknown genes specifically or highly expressed in the brain of the Henan gamecock were identified. These genes are potentially key candidates for enhancing the bird's aggression. Multi-omics joint analysis revealed that tyrosine metabolism and neuroactive ligand-receptor interaction were commonly affected. Overall, our results propose that the aggressiveness of Henan gamecocks can be heightened by the activation of the serotonergic-dopaminergic metabolic process in the brain, which concurrently impairs the neuroimmune system. Further research is needed to identify the function of these unknown genes on the bird's aggressive behavior.
Collapse
Affiliation(s)
- Chuanchen Su
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Lin Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Yuxian Pan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Jingya Jiao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Pengna Luo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Xinghai Chang
- Henan Changxing Agriculture and Animal Husbandry co., LTD, Kaifeng, Henan 475000, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Xuemeng Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Wen Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China
| | - Yanqun Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou Henan 450046, China.
| |
Collapse
|
6
|
Tao L, Ayambem D, Barranca VJ, Bhandawat V. Neurons Underlying Aggression-Like Actions That Are Shared by Both Males and Females in Drosophila. J Neurosci 2024; 44:e0142242024. [PMID: 39317475 PMCID: PMC11529818 DOI: 10.1523/jneurosci.0142-24.2024] [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: 01/20/2024] [Revised: 09/05/2024] [Accepted: 09/11/2024] [Indexed: 09/26/2024] Open
Abstract
Aggression involves both sexually monomorphic and dimorphic actions. How the brain implements these two types of actions is poorly understood. We found that in Drosophila melanogaster, a set of neurons, which we call CL062, previously shown to mediate male aggression also mediate female aggression. These neurons elicit aggression acutely and without the presence of a target. Although the same set of actions is elicited in males and females, the overall behavior is sexually dimorphic. The CL062 neurons do not express fruitless, a gene required for sexual dimorphism in flies, and expressed by most other neurons important for controlling fly aggression. Connectomic analysis in a female electron microscopy dataset suggests that these neurons have limited connections with fruitless expressing neurons that have been shown to be important for aggression and signal to different descending neurons. Thus, CL062 is part of a monomorphic circuit for aggression that functions parallel to the known dimorphic circuits.
Collapse
Affiliation(s)
- Liangyu Tao
- School of Biomedical Engineering and Health Sciences, Drexel University, Philadelphia, Pennsylvania 19104
| | | | | | - Vikas Bhandawat
- School of Biomedical Engineering and Health Sciences, Drexel University, Philadelphia, Pennsylvania 19104
| |
Collapse
|
7
|
Jackson LR, Alward BA. Sexually dimorphic control of aggression by androgen signaling in a cichlid. Mol Cell Endocrinol 2024; 592:112319. [PMID: 38925266 PMCID: PMC11446503 DOI: 10.1016/j.mce.2024.112319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Innate social behaviors like aggression are modulated by sex steroid hormones such as androgens and estrogens. However, we know little about how the same hormone regulates similar behaviors in both sexes. We investigated the role of androgenic signaling in the regulation of aggression in Astatotilapia burtoni, a social fish in which males and females perform similar aggressive behaviors. We used androgen receptor (AR) α knockout (KO) animals for this study since this gene was recently shown to be required for male-typical aggression and mating. Surprisingly, ARα KO females did not show deficits in aggression. We also determined that females lacking the other AR, ARβ, showed normal levels of aggression. Blocking both ARs pharmacologically confirmed that neither AR is necessary for aggression in females. However, ARα KO males showed clear deficits in attacks. Thus, in A. burtoni there appears to be a sexual dimorphism in the role of ARα in the control of aggression.
Collapse
Affiliation(s)
| | - Beau A Alward
- University of Houston, Department of Psychology, USA; University of Houston, Department of Biology and Biochemistry, USA; University of California, Los Angeles, Department of Integrative Biology and Physiology, USA.
| |
Collapse
|
8
|
Zhu Z, Miao L, Li K, Ma Q, Pan L, Shen C, Ge Q, Du Y, Yin L, Yang H, Xu X, Zeng LH, Liu Y, Xu H, Li XM, Sun L, Yu YQ, Duan S. A hypothalamic-amygdala circuit underlying sexually dimorphic aggression. Neuron 2024; 112:3176-3191.e7. [PMID: 39019042 DOI: 10.1016/j.neuron.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 05/13/2024] [Accepted: 06/20/2024] [Indexed: 07/19/2024]
Abstract
Male animals often display higher levels of aggression than females. However, the neural circuitry mechanisms underlying this sexually dimorphic aggression remain elusive. Here, we identify a hypothalamic-amygdala circuit that mediates male-biased aggression in mice. Specifically, the ventrolateral part of the ventromedial hypothalamus (VMHvl), a sexually dimorphic region associated with eliciting male-biased aggression, projects densely to the posterior substantia innominata (pSI), an area that promotes similar levels of attack in both sexes of mice. Although the VMHvl innervates the pSI unidirectionally through both excitatory and inhibitory connections, it is the excitatory VMHvl-pSI projections that are strengthened in males to promote aggression, whereas the inhibitory connections that reduce aggressive behavior are strengthened in females. Consequently, the convergent hypothalamic input onto the pSI leads to heightened pSI activity in males, resulting in male-biased aggression. Our findings reveal a sexually distinct excitation-inhibition balance of a hypothalamic-amygdala circuit that underlies sexually dimorphic aggression.
Collapse
Affiliation(s)
- Zhenggang Zhu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Lu Miao
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Kaiyuan Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Qingqing Ma
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Lina Pan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Chenjie Shen
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Qianqian Ge
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yonglan Du
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Luping Yin
- Westlake Laboratory of Life Sciences and Biomedicine, Institute of Biology, School of Life Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, China
| | - Hongbin Yang
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Xiaohong Xu
- Institute of Neuroscience and Key Laboratory of Primate Neurobiology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Yijun Liu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Han Xu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao-Ming Li
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Li Sun
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Yan-Qin Yu
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China.
| | - Shumin Duan
- Department of Neurology of Second Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science & Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou 311121, China; NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China; Research Units for Emotion and Emotion Disorders, Chinese Academy of Medical Sciences, Hangzhou, China.
| |
Collapse
|
9
|
Singh R, Gobrogge K. Aggression Unleashed: Neural Circuits from Scent to Brain. Brain Sci 2024; 14:794. [PMID: 39199486 PMCID: PMC11352925 DOI: 10.3390/brainsci14080794] [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: 06/18/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Aggression is a fundamental behavior with essential roles in dominance assertion, resource acquisition, and self-defense across the animal kingdom. However, dysregulation of the aggression circuitry can have severe consequences in humans, leading to economic, emotional, and societal burdens. Previous inconsistencies in aggression research have been due to limitations in techniques for studying these neurons at a high spatial resolution, resulting in an incomplete understanding of the neural mechanisms underlying aggression. Recent advancements in optogenetics, pharmacogenetics, single-cell RNA sequencing, and in vivo electrophysiology have provided new insights into this complex circuitry. This review aims to explore the aggression-provoking stimuli and their detection in rodents, particularly through the olfactory systems. Additionally, we will examine the core regions associated with aggression, their interactions, and their connection with the prefrontal cortex. We will also discuss the significance of top-down cognitive control systems in regulating atypical expressions of aggressive behavior. While the focus will primarily be on rodent circuitry, we will briefly touch upon the modulation of aggression in humans through the prefrontal cortex and discuss emerging therapeutic interventions that may benefit individuals with aggression disorders. This comprehensive understanding of the neural substrates of aggression will pave the way for the development of novel therapeutic strategies and clinical interventions. This approach contrasts with the broader perspective on neural mechanisms of aggression across species, aiming for a more focused analysis of specific pathways and their implications for therapeutic interventions.
Collapse
Affiliation(s)
- Rhea Singh
- The Mortimer B. Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Columbia University, New York, NY 10027, USA
| | - Kyle Gobrogge
- Undergraduate Program in Neuroscience, Boston University, Boston, MA 02215, USA;
| |
Collapse
|
10
|
Sofer Y, Zilkha N, Gimpel E, Wagner S, Chuartzman SG, Kimchi T. Sexually dimorphic oxytocin circuits drive intragroup social conflict and aggression in wild house mice. Nat Neurosci 2024; 27:1565-1573. [PMID: 38969756 DOI: 10.1038/s41593-024-01685-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 05/16/2024] [Indexed: 07/07/2024]
Abstract
In nature, both males and females engage in competitive aggressive interactions to resolve social conflicts, yet the behavioral principles guiding such interactions and their underlying neural mechanisms remain poorly understood. Through circuit manipulations in wild mice, we unveil oxytocin-expressing (OT+) neurons in the hypothalamic paraventricular nucleus (PVN) as a neural hub governing behavior in dyadic and intragroup social conflicts, influencing the degree of behavioral sexual dimorphism. We demonstrate that OT+ PVN neurons are essential and sufficient in promoting aggression and dominance hierarchies, predominantly in females. Furthermore, pharmacogenetic activation of these neurons induces a change in the 'personality' traits of the mice within groups, in a sex-dependent manner. Finally, we identify an innervation from these OT neurons to the ventral tegmental area that drives dyadic aggression, in a sex-specific manner. Our data suggest that competitive aggression in naturalistic settings is mediated by a sexually dimorphic OT network connected with reward-related circuitry.
Collapse
Affiliation(s)
- Yizhak Sofer
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Noga Zilkha
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Elena Gimpel
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomo Wagner
- Sagol Department of Neurobiology, the Integrated Brain and Behavior Research Center, University of Haifa, Haifa, Israel
| | | | - Tali Kimchi
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
11
|
Chen H, Ferguson CJ, Mitchell DC, Titus A, Paulo JA, Hwang A, Lin TH, Yano H, Gu W, Song SK, Yuede CM, Gygi SP, Bonni A, Kim AH. The Hao-Fountain syndrome protein USP7 regulates neuronal connectivity in the brain via a novel p53-independent ubiquitin signaling pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.24.563880. [PMID: 37961719 PMCID: PMC10634808 DOI: 10.1101/2023.10.24.563880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Precise control of protein ubiquitination is essential for brain development, and hence, disruption of ubiquitin signaling networks can lead to neurological disorders. Mutations of the deubiquitinase USP7 cause the Hao-Fountain syndrome (HAFOUS), characterized by developmental delay, intellectual disability, autism, and aggressive behavior. Here, we report that conditional deletion of USP7 in excitatory neurons in the mouse forebrain triggers diverse phenotypes including sensorimotor deficits, learning and memory impairment, and aggressive behavior, resembling clinical features of HAFOUS. USP7 deletion induces neuronal apoptosis in a manner dependent of the tumor suppressor p53. However, most behavioral abnormalities in USP7 conditional mice persist despite p53 loss. Strikingly, USP7 deletion in the brain perturbs the synaptic proteome and dendritic spine morphogenesis independently of p53. Integrated proteomics analysis reveals that the neuronal USP7 interactome is enriched for proteins implicated in neurodevelopmental disorders and specifically identifies the RNA splicing factor Ppil4 as a novel neuronal substrate of USP7. Knockdown of Ppil4 in cortical neurons impairs dendritic spine morphogenesis, phenocopying the effect of USP7 loss on dendritic spines. These findings reveal a novel USP7-Ppil4 ubiquitin signaling link that regulates neuronal connectivity in the developing brain, with implications for our understanding of the pathogenesis of HAFOUS and other neurodevelopmental disorders.
Collapse
|
12
|
Lin JC, Daigle CL, Tang PC, Wang CK. Influence of sex hormones on the aggressive behavior during peck order establishment and stabilization in meat and egg type chickens. Poult Sci 2024; 103:103669. [PMID: 38603931 PMCID: PMC11017360 DOI: 10.1016/j.psj.2024.103669] [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: 12/06/2023] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
In the poultry industry, broiler and layer strains are genetically selected for different purposes (e.g., high meat-yield and high egg-production). Genetic selection for productivity can have unintended consequences on the behavioral repertoire of the birds, including aggression. Alongside the increasing societal concern regarding the welfare of animal in agriculture, the number of countries that are advocating the prohibition of using battery cages for laying hens has resulted in the transition and adoption of cage-free or free-range systems. Thus, both broiler and layer chickens are housed in large flocks rather than housed individually in cages. Housing birds in groups increases the opportunity for birds to engage in social behaviors, including aggression, that are used to establish social status. Aggressive interactions are associated with the risk of injury and the potential for a subordinate animal to have unmet needs (e.g., access to feed). The aim of this study was to characterize the relationships among aggressive behavior, neurobiology, and hormones during peck order establishment and social hierarchy stabilization of 2 divergently selected strains (meat- and egg-type chicken). Meat-type strains performed more male on male (P < 0.001), male on female (P < 0.0001), and female on female (P < 0.0001) non-reciprocal aggression behavior (NRA) than egg-type strains. Greater serum testosterone and estradiol concentrations in the weeks after the peck order establishment were observed in meat-type birds compared those in egg-type birds for both males and females (all P < 0.05). Greater (P < 0.05) cellular densities of androgen receptors, but not estrogen receptors, were observed in the hypothalamus of meat-type birds compared to egg-type birds. These findings suggest that greater sex hormone concentrations in the meat-type birds may be a consequence of genetic selection for rapid growth resulting in more sex hormones-induced aggressive behavior.
Collapse
Affiliation(s)
- Jou-Ching Lin
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Courtney Lynd Daigle
- Department of Animal Science, Texas A&M University, Kleberg Center, College Station, TX 77843, USA
| | - Pin-Chi Tang
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Chien-Kai Wang
- Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan; The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
13
|
Rogers JF, Vandendoren M, Prather JF, Landen JG, Bedford NL, Nelson AC. Neural cell-types and circuits linking thermoregulation and social behavior. Neurosci Biobehav Rev 2024; 161:105667. [PMID: 38599356 PMCID: PMC11163828 DOI: 10.1016/j.neubiorev.2024.105667] [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: 01/03/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Understanding how social and affective behavioral states are controlled by neural circuits is a fundamental challenge in neurobiology. Despite increasing understanding of central circuits governing prosocial and agonistic interactions, how bodily autonomic processes regulate these behaviors is less resolved. Thermoregulation is vital for maintaining homeostasis, but also associated with cognitive, physical, affective, and behavioral states. Here, we posit that adjusting body temperature may be integral to the appropriate expression of social behavior and argue that understanding neural links between behavior and thermoregulation is timely. First, changes in behavioral states-including social interaction-often accompany changes in body temperature. Second, recent work has uncovered neural populations controlling both thermoregulatory and social behavioral pathways. We identify additional neural populations that, in separate studies, control social behavior and thermoregulation, and highlight their relevance to human and animal studies. Third, dysregulation of body temperature is linked to human neuropsychiatric disorders. Although body temperature is a "hidden state" in many neurobiological studies, it likely plays an underappreciated role in regulating social and affective states.
Collapse
Affiliation(s)
- Joseph F Rogers
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Morgane Vandendoren
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Jonathan F Prather
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Jason G Landen
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA
| | - Nicole L Bedford
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA
| | - Adam C Nelson
- Department of Zoology & Physiology, University of Wyoming, Laramie, WY, USA; University of Wyoming Sensory Biology Center, USA.
| |
Collapse
|
14
|
Johnson MC, Zweig JA, Zhang Y, Ryabinin AE. Effects of social housing on alcohol intake in mice depend on the non-social environment. Front Behav Neurosci 2024; 18:1380031. [PMID: 38817806 PMCID: PMC11137225 DOI: 10.3389/fnbeh.2024.1380031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/01/2024] [Indexed: 06/01/2024] Open
Abstract
Background Excessive alcohol consumption leads to serious health problems. Mechanisms regulating the consumption of alcohol are insufficiently understood. Previous preclinical studies suggested that non-social environmental and social environmental complexities can regulate alcohol consumption in opposite directions. However, previous studies did not include all conditions and/or did not include female rodents. Therefore, in this study, we examined the effects of social versus single housing in standard versus non-standard housing conditions in male and female mice. Methods Adult C57BL/6 J mice were housed in either standard shoebox cages or in automated Herdsman 2 (HM2) cages and exposed to a two-bottle choice procedure with 3% or 6% ethanol versus water for 5 days. The HM2 cages use radiotracking devices to measure the fluid consumption of individual mice in an undisturbed and automated manner. In both housing conditions, mice were housed either at one or at four per cage. Results In standard cages, group housing of animals decreased alcohol consumption and water consumption. In HM2 cages, group housing significantly increased ethanol preference and decreased water intake. There were no significant differences in these effects between male and female animals. These observations were similar for 3 and 6% ethanol solutions but were more pronounced for the latter. The effects of social environment on ethanol preference in HM2 cages were accompanied by an increase in the number of approaches to the ethanol solution and a decrease in the number of approaches to water. The differences in ethanol intake could not be explained by differences in locomotor or exploratory activity as socially housed mice showed fewer non-consummatory visits to the ethanol solutions than single-housed animals. In addition, we observed that significant changes in behaviors measuring the approach to the fluid were not always accompanied by significant changes in fluid consumption, and vice versa, suggesting that it is important to assess both measures of motivation to consume alcohol. Conclusion Our results indicate that the direction of the effects of social environment on alcohol intake in mice depends on the non-social housing environment. Understanding mechanisms by which social and non-social housing conditions modulate alcohol intake could suggest approaches to counteract environmental factors enhancing hazardous alcohol consumption.
Collapse
Affiliation(s)
| | | | | | - Andrey E. Ryabinin
- Department of Behavioral Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
15
|
Jackson LR, Alward B. Sexually dimorphic control of aggression by androgen signaling in a cichlid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587979. [PMID: 38617319 PMCID: PMC11014533 DOI: 10.1101/2024.04.03.587979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Innate social behaviors like aggression are modulated by sex steroid hormones such as androgens and estrogens. However, we know little about how the same hormone regulates similar behaviors in both sexes. We investigated the role of androgenic signaling in the regulation of aggression in Astatotilapia burtoni, a social fish in which males and females perform similar aggressive behaviors. We used ARa knockout (KO) animals for this study, which was recently shown to be required for male-typical aggression and mating. Surprisingly, ARα KO females did not show deficits in aggression. We also determined that females lacking the other AR, ARβ, showed normal levels of aggression. Blocking both ARs pharmacologically confirmed that neither AR is necessary for aggression in females. However, ARα KO males showed clear deficits in attacks. Thus, in A. burtoni there appears to be a sexual dimorphism in the role of ARα in the control of aggression.
Collapse
Affiliation(s)
| | - Beau Alward
- University of Houston, Department of Psychology
- University of Houston, Department of Biology and Biochemistry
| |
Collapse
|
16
|
Minakuchi T, Guthman EM, Acharya P, Hinson J, Fleming W, Witten IB, Oline SN, Falkner AL. Independent inhibitory control mechanisms for aggressive motivation and action. Nat Neurosci 2024; 27:702-715. [PMID: 38347201 DOI: 10.1038/s41593-023-01563-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/19/2023] [Indexed: 04/10/2024]
Abstract
Social behaviors often consist of a motivational phase followed by action. Here we show that neurons in the ventromedial hypothalamus ventrolateral area (VMHvl) of mice encode the temporal sequence of aggressive motivation to action. The VMHvl receives local inhibitory input (VMHvl shell) and long-range input from the medial preoptic area (MPO) with functional coupling to neurons with specific temporal profiles. Encoding models reveal that during aggression, VMHvl shellvgat+ activity peaks at the start of an attack, whereas activity from the MPO-VMHvlvgat+ input peaks at specific interaction endpoints. Activation of the MPO-VMHvlvgat+ input promotes and prolongs a low motivation state, whereas activation of VMHvl shellvgat+ results in action-related deficits, acutely terminating attack. Moreover, stimulation of MPO-VMHvlvgat+ input is positively valenced and anxiolytic. Together, these data demonstrate how distinct inhibitory inputs to the hypothalamus can independently gate the motivational and action phases of aggression through a single locus of control.
Collapse
Affiliation(s)
| | | | | | - Justin Hinson
- Princeton Neuroscience Institute, Princeton, NJ, USA
| | | | | | | | | |
Collapse
|
17
|
Tao L, Ayembem D, Barranca VJ, Bhandawat V. Neurons underlying aggressive actions that are shared by both males and females in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.26.582148. [PMID: 38464020 PMCID: PMC10925114 DOI: 10.1101/2024.02.26.582148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Aggression involves both sexually monomorphic and dimorphic actions. How the brain implements these two types of actions is poorly understood. We found that a set of neurons, which we call CL062, previously shown to mediate male aggression also mediate female aggression. These neurons elicit aggression acutely and without the presence of a target. Although the same set of actions is elicited in males and females, the overall behavior is sexually dimorphic. The CL062 neurons do not express fruitless , a gene required for sexual dimorphism in flies, and expressed by most other neurons important for controlling fly aggression. Connectomic analysis suggests that these neurons have limited connections with fruitless expressing neurons that have been shown to be important for aggression, and signal to different descending neurons. Thus, CL062 is part of a monomorphic circuit for aggression that functions parallel to the known dimorphic circuits.
Collapse
|
18
|
Kawabata-Sakata Y, Kanda S, Okubo K. Male-specific vasotocin expression in the medaka tuberal hypothalamus: Androgen dependence and probable role in aggression. Mol Cell Endocrinol 2024; 580:112101. [PMID: 37923055 DOI: 10.1016/j.mce.2023.112101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
Terrestrial vertebrates have a population of androgen-dependent vasotocin (VT)-expressing neurons in the extended amygdala that are more abundant in males and mediate male-typical social behaviors, including aggression. Teleosts lack these neurons but instead have novel male-specific VT-expressing neurons in the tuberal hypothalamus. Here we found in medaka that vt expression in these neurons is dependent on post-pubertal gonadal androgens and that androgens can act on these neurons to directly stimulate vt transcription via the androgen receptor subtype Ara. Furthermore, administration of exogenous VT induced aggression in females and alterations in the androgen milieu led to correlated changes in the levels of tuberal hypothalamic vt expression and aggression in both sexes. However, genetic ablation of vt failed to prevent androgen-induced aggression in females. Collectively, our results demonstrate a marked androgen dependence of male-specific vt expression in the teleost tuberal hypothalamus, although its relevance to male-typical aggression needs to be further validated.
Collapse
Affiliation(s)
- Yukika Kawabata-Sakata
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan; Department of Pathophysiology, Tokyo Medical University, Shinjuku, Tokyo, 160-8402, Japan
| | - Shinji Kanda
- Laboratory of Physiology, Atmosphere and Ocean Research Institute, The University of Tokyo, Kashiwa, Chiba, 277-8564, Japan
| | - Kataaki Okubo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, 113-8657, Japan.
| |
Collapse
|
19
|
Mei L, Osakada T, Lin D. Hypothalamic control of innate social behaviors. Science 2023; 382:399-404. [PMID: 37883550 PMCID: PMC11105421 DOI: 10.1126/science.adh8489] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
Sexual, parental, and aggressive behaviors are central to the reproductive success of individuals and species survival and thus are supported by hardwired neural circuits. The reproductive behavior control column (RBCC), which comprises the medial preoptic nucleus (MPN), the ventrolateral part of the ventromedial hypothalamus (VMHvl), and the ventral premammillary nucleus (PMv), is essential for all social behaviors. The RBCC integrates diverse hormonal and metabolic cues and adjusts an animal's physical activity, hence the chance of social encounters. The RBCC further engages the mesolimbic dopamine system to maintain social interest and reinforces cues and actions that are time-locked with social behaviors. We propose that the RBCC and brainstem form a dual-control system for generating moment-to-moment social actions. This Review summarizes recent progress regarding the identities of RBCC cells and their pathways that drive different aspects of social behaviors.
Collapse
Affiliation(s)
- Long Mei
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Takuya Osakada
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Dayu Lin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
- Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, NY 10016, USA
- Center for Neural Science, New York University, New York, NY 10016, USA
| |
Collapse
|
20
|
Abstract
Rapid advances in the neural control of social behavior highlight the role of interconnected nodes engaged in differential information processing to generate behavior. Many innate social behaviors are essential to reproductive fitness and therefore fundamentally different in males and females. Programming these differences occurs early in development in mammals, following gonadal differentiation and copious androgen production by the fetal testis during a critical period. Early-life programming of social behavior and its adult manifestation are separate but yoked processes, yet how they are linked is unknown. This review seeks to highlight that gap by identifying four core mechanisms (epigenetics, cell death, circuit formation, and adult hormonal modulation) that could connect developmental changes to the adult behaviors of mating and aggression. We further propose that a unique social behavior, adolescent play, bridges the preweaning to the postpubertal brain by engaging the same neural networks underpinning adult reproductive and aggressive behaviors.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland, USA;
| |
Collapse
|
21
|
Xiang J, Guo RY, Wang T, Zhang N, Chen XR, Li EC, Zhang JL. Brain metabolite profiles provide insight into mechanisms for behavior sexual dimorphisms in zebrafish (Danio rerio). Physiol Behav 2023; 263:114132. [PMID: 36801416 DOI: 10.1016/j.physbeh.2023.114132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/27/2022] [Accepted: 02/16/2023] [Indexed: 02/19/2023]
Abstract
The zebrafish (Danio rerio) has historically been a useful model for research in genetics, ecology, biology, toxicology, and neurobehavior. Zebrafish have been demonstrated to have brain sexual dimorphism. However, the sexual dimorphism of zebrafish behavior demands our attention, particularly. To evaluate the behavior and brain sexual dimorphisms in zebrafish, this study assessed sex differences in adult D. rerio in four behavioral domains, including aggression, fear, anxiety, and shoaling, and further compared with metabolites in the brain tissue of females and males. Our findings showed that aggression, fear, anxiety and shoaling behaviors were significantly sexually dimorphic. Interestingly, we also show through a novel data analysis method, that the female zebrafish exhibited significantly increased shoaling behavior when shoaled with male zebrafish groups and, for the first time, we offer evidence that male shoals are beneficial in dramatically alleviating anxiety in zebrafish. In addition, there were significant changes in metabolites in zebrafish brain tissue between the sexes. Furthermore, zebrafish behavioral sexual dimorphism may be associated with brain sexual dimorphism, with significant differences in brain metabolites. Therefore, to prevent the influence or even bias of behavioral sex differences on results, it is suggested that behavioral studies or behavioral-based other relevant investigations consider sexual dimorphism of behavior and brain.
Collapse
Affiliation(s)
- Jing Xiang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Rui-Ying Guo
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Ting Wang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Nan Zhang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Xian-Rui Chen
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China
| | - Er-Chao Li
- College of Marine Sciences, Hainan University, Haikou, Hainan, China
| | - Ji-Liang Zhang
- Ministry of Education Key Laboratory for Ecology of Tropical Islands, Key Laboratory of Tropical Animal and Plant Ecology of Hainan Province, College of Life Sciences, Hainan Normal University, Haikou, Hainan, China.
| |
Collapse
|
22
|
Yang J, Liu Y, Fan Y, Shen D, Shen J, Fang G. High-Frequency Local Field Potential Oscillations May Modulate Aggressive Behaviors in Mice. BIOLOGY 2022; 11:1682. [PMID: 36421396 PMCID: PMC9687601 DOI: 10.3390/biology11111682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 07/03/2024]
Abstract
Aggressive behavior is one of congenital social behaviors in many species, which could be promoted by social neglect or isolation in the early stages of life. Many brain regions including the medial prefrontal cortex (mPFC), medial amygdala (MeA) and ventromedial hypothalamus (VMH) are demonstrated to relate to aggressive behavior; however, the dynamic patterns of neural activities during the occurrence of this behavior remain unclear. In this study, 21-day-old male CD-1 mice were reared in social isolation conditions and cohousing conditions for two weeks. Aggressive behaviors of each subject were estimated by the resident-intruder test. Simultaneously, the local field potentials of mPFC, MeA and VMH were recorded for exploring differences in the relative power spectra of different oscillations when aggressive behaviors occurred. The results showed that the following: (1) Compared with the cohousing mice, the socially isolated mice exhibited more aggression. (2) Regardless of "time condition" (pre-, during- and post- attack), the relative power spectra of beta band in the cohousing mice were significantly greater than those in the socially isolated mice, and inversely, the relative power spectra of gamma band in the cohousing mice were significantly smaller than those in the socially isolated mice. (3) The bilateral mPFC exhibited significantly smaller beta power spectra but greater gamma power spectra compared with other brain areas regardless of rearing patterns. (4) For the right VMH of the socially isolated mice, the relative power spectra of the gamma band during attacks were significantly greater than those before attack. These results suggest that aggressive behaviors in mice could be shaped by rearing patterns and that high-frequency oscillations (beta and gamma bands) may engage in mediating aggressive behaviors in mice.
Collapse
Affiliation(s)
- Jing Yang
- Chengdu Institute of Biology, Chinese Academy of Sciences, No.9 Section 4, Renmin Nan Road, Chengdu 610041, China
| | - Yansu Liu
- Sichuan Nursing Vocational College, No. 173 Longdu Nan Road, Chengdu 610100, China
| | - Yanzhu Fan
- Chengdu Institute of Biology, Chinese Academy of Sciences, No.9 Section 4, Renmin Nan Road, Chengdu 610041, China
| | - Di Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, No.9 Section 4, Renmin Nan Road, Chengdu 610041, China
| | - Jiangyan Shen
- Chengdu Institute of Biology, Chinese Academy of Sciences, No.9 Section 4, Renmin Nan Road, Chengdu 610041, China
| | - Guangzhan Fang
- Chengdu Institute of Biology, Chinese Academy of Sciences, No.9 Section 4, Renmin Nan Road, Chengdu 610041, China
- Key Laboratory of Southwest China Wildlife Resources Conservation (Ministry of Education), China West Normal University, No. 1 Shi Da Road, Nanchong 637009, China
| |
Collapse
|
23
|
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
|
24
|
Parker CG, Lee JS, Histed AR, Craig SE, Rhodes JS. Stable and persistent male-like behavior during male-to-female sex change in the common clownfish Amphiprion ocellaris. Horm Behav 2022; 145:105239. [PMID: 35926412 DOI: 10.1016/j.yhbeh.2022.105239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 07/04/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
Many fish species exhibit natural sex change as part of their life, providing unique opportunities to study sexually-differentiated social behaviors and their plasticity. Past research has shown that behavioral sex change in the female-to-male (protogynous) direction occurs rapidly and well before gonadal sex change. However, little is known about the timecourse of behavioral sex change in male-to-female (protandrous) sex-changing species, limiting our ability to compare patterns of behavioral sex change across species and identify conserved or divergent underlying mechanisms. Using the protandrous sex changing anemonefish Amphiprion ocellaris, we assessed behavior (aggression and parental care) and hormones (estradiol and 11-ketotestosterone) in fish over six months of sex change, and compared those fish against their non-changing partners as well as control males and females. Contrary to expectations, we found that sex-changing fish displayed behavior that was persistently male-like, and that their behavior did not become progressively female-like as sex change progressed. Hormones shifted to an intermediate profile between males and females and remained stable until gonads changed. These results support a new perspective that the timecourse for protandrous sex change in anemonefish is completely distinct from other well-established models, such that behavioral sex change does not occur until after gonadal sex change is complete, and that sex-changing fish have a stable and unique behavioral and hormonal phenotype that is distinct from a male-typical or female-typical phenotype. The results also identify aspects of sex change that may fundamentally differ between protandrous and protogynous modes, motivating further research into these remarkable examples of phenotypic plasticity.
Collapse
Affiliation(s)
- Coltan G Parker
- Neuroscience Program, University of Illinois, Urbana-Champaign, IL, USA
| | - Joanne S Lee
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, USA
| | - Abigail R Histed
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, USA
| | - Sarah E Craig
- Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, USA
| | - Justin S Rhodes
- Neuroscience Program, University of Illinois, Urbana-Champaign, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, USA; Department of Psychology, University of Illinois, Urbana-Champaign, IL, USA.
| |
Collapse
|
25
|
Pechuk V, Goldman G, Salzberg Y, Chaubey AH, Bola RA, Hoffman JR, Endreson ML, Miller RM, Reger NJ, Portman DS, Ferkey DM, Schneidman E, Oren-Suissa M. Reprogramming the topology of the nociceptive circuit in C. elegans reshapes sexual behavior. Curr Biol 2022; 32:4372-4385.e7. [PMID: 36075218 DOI: 10.1016/j.cub.2022.08.038] [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: 02/09/2022] [Revised: 06/28/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
The effect of the detailed connectivity of a neural circuit on its function and the resulting behavior of the organism is a key question in many neural systems. Here, we study the circuit for nociception in C. elegans, which is composed of the same neurons in the two sexes that are wired differently. We show that the nociceptive sensory neurons respond similarly in the two sexes, yet the animals display sexually dimorphic behaviors to the same aversive stimuli. To uncover the role of the downstream network topology in shaping behavior, we learn and simulate network models that replicate the observed dimorphic behaviors and use them to predict simple network rewirings that would switch behavior between the sexes. We then show experimentally that these subtle synaptic rewirings indeed flip behavior. Interestingly, when presented with aversive cues, rewired males were compromised in finding mating partners, suggesting that network topologies that enable efficient avoidance of noxious cues have a reproductive "cost." Our results present a deconstruction of the design of a neural circuit that controls sexual behavior and how to reprogram it.
Collapse
Affiliation(s)
- Vladyslava Pechuk
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gal Goldman
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yehuda Salzberg
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Aditi H Chaubey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - R Aaron Bola
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Jonathon R Hoffman
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Morgan L Endreson
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Renee M Miller
- Department of Brain and Cognitive Sciences, University of Rochester, Rochester, NY 14627, USA
| | - Noah J Reger
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA
| | - Douglas S Portman
- Department of Biomedical Genetics, University of Rochester, Rochester, NY 14642, USA
| | - Denise M Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Elad Schneidman
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Meital Oren-Suissa
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
26
|
Dai B, Sun F, Tong X, Ding Y, Kuang A, Osakada T, Li Y, Lin D. Responses and functions of dopamine in nucleus accumbens core during social behaviors. Cell Rep 2022; 40:111246. [PMID: 36001967 PMCID: PMC9511885 DOI: 10.1016/j.celrep.2022.111246] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 04/22/2022] [Accepted: 07/31/2022] [Indexed: 12/05/2022] Open
Abstract
Social behaviors are among the most important motivated behaviors. How dopamine (DA), a "reward" signal, releases during social behaviors has been a topic of interest for decades. Here, we use a genetically encoded DA sensor, GRABDA2m, to record DA activity in the nucleus accumbens (NAc) core during various social behaviors in male and female mice. We find that DA releases during approach, investigation and consummation phases of social behaviors signal animals' motivation, familiarity of the social target, and valence of the experience, respectively. Positive and negative social experiences evoke opposite DA patterns. Furthermore, DA releases during mating and fighting are sexually dimorphic with a higher level in males than in females. At the functional level, increasing DA in NAc enhances social interest toward a familiar conspecific and alleviates defeat-induced social avoidance. Altogether, our results reveal complex information encoded by NAc DA activity during social behaviors and their multistage functional roles.
Collapse
Affiliation(s)
- Bing Dai
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA.
| | - Fangmiao Sun
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Xiaoyu Tong
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Yizhuo Ding
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Amy Kuang
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Takuya Osakada
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China; PKU-IDG/McGovern Institute for Brain Research, Beijing, China; Peking-Tsinghua Center for Life Sciences, Beijing, China
| | - Dayu Lin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
27
|
Oliveira VEDM, de Jong TR, Neumann ID. Synthetic Oxytocin and Vasopressin Act Within the Central Amygdala to Exacerbate Aggression in Female Wistar Rats. Front Neurosci 2022; 16:906617. [PMID: 35663559 PMCID: PMC9158429 DOI: 10.3389/fnins.2022.906617] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/28/2022] [Indexed: 11/15/2022] Open
Abstract
Exacerbated aggression is a high-impact, but poorly understood core symptom of several psychiatric disorders, which can also affect women. Animal models have successfully been employed to unravel the neurobiology of aggression. However, despite increasing evidence for sex-specificity, little is known about aggression in females. Here, we studied the role of the oxytocin (OXT) and arginine vasopressin (AVP) systems within the central amygdala (CeA) on aggressive behavior displayed by virgin female Wistar rats using immunohistochemistry, receptor autoradiography, and neuropharmacology. Our data show that CeA GABAergic neurons are activated after an aggressive encounter in the female intruder test. Additionally, neuronal activity (pERK) negatively correlated with the display of aggression in low-aggressive group-housed females. Binding of OXT receptors, but not AVP-V1a receptors, was increased in the CeA of high-aggressive isolated and trained (IST) females. Finally, local infusion of either synthetic OXT or AVP enhanced aggression in IST females, whereas blockade of either of these receptors did not affect aggressive behavior. Altogether, our data support a moderate role of the CeA in female aggression. Regarding neuropeptide signaling, our findings suggest that synthetic, but not endogenous OXT and AVP modulate aggressive behavior in female Wistar rats.
Collapse
Affiliation(s)
- Vinícius E. de M. Oliveira
- Laboratory of Neuroendocrinology, GIGA-Neurosciences, University of Liege, Liege, Belgium
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Trynke R. de Jong
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
- Medische Biobank Noord-Nederland B.V., Groningen, Netherlands
| | - Inga D. Neumann
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
- *Correspondence: Inga D. Neumann,
| |
Collapse
|
28
|
Reichmann F, Pilic J, Trajanoski S, Norton WHJ. Transcriptomic underpinnings of high and low mirror aggression zebrafish behaviours. BMC Biol 2022; 20:97. [PMID: 35501893 PMCID: PMC9059464 DOI: 10.1186/s12915-022-01298-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 04/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Aggression is an adaptive behaviour that animals use to protect offspring, defend themselves and obtain resources. Zebrafish, like many other animals, are not able to recognize themselves in the mirror and typically respond to their own reflection with aggression. However, mirror aggression is not an all-or-nothing phenomenon, with some individuals displaying high levels of aggression against their mirror image, while others show none at all. In the current work, we have investigated the genetic basis of mirror aggression by using a classic forward genetics approach - selective breeding for high and low mirror aggression zebrafish (HAZ and LAZ). Results We characterized AB wild-type zebrafish for their response to the mirror image. Both aggressive and non-aggressive fish were inbred over several generations. We found that HAZ were on average more aggressive than the corresponding LAZ across generations and that the most aggressive adult HAZ were less anxious than the least aggressive adult LAZ after prolonged selective breeding. RNAseq analysis of these fish revealed that hundreds of protein-encoding genes with important diverse biological functions such as arsenic metabolism (as3mt), cell migration (arl4ab), immune system activity (ptgr1), actin cytoskeletal remodelling (wdr1), corticogenesis (dgcr2), protein dephosphorylation (ublcp1), sialic acid metabolism (st6galnac3) and ketone body metabolism (aacs) were differentially expressed between HAZ and LAZ, suggesting a strong genetic contribution to this phenotype. DAVID pathway analysis showed that a number of diverse pathways are enriched in HAZ over LAZ including pathways related to immune function, oxidation-reduction processes and cell signalling. In addition, weighted gene co-expression network analysis (WGCNA) identified 12 modules of highly correlated genes that were significantly associated with aggression duration and/or experimental group. Conclusions The current study shows that selective breeding based of the mirror aggression phenotype induces strong, heritable changes in behaviour and gene expression within the brain of zebrafish suggesting a strong genetic basis for this behaviour. Our transcriptomic analysis of fish selectively bred for high and low levels of mirror aggression revealed specific transcriptomic signatures induced by selective breeding and mirror aggression and thus provides a large and novel resource of candidate genes for future study. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01298-z.
Collapse
Affiliation(s)
- Florian Reichmann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
| | - Johannes Pilic
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Slave Trajanoski
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - William H J Norton
- Department of Genetics and Genome Biology, College of Life Sciences, University of Leicester, Leicester, UK. .,Department of Genetics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
29
|
Neural mechanisms of persistent aggression. Curr Opin Neurobiol 2022; 73:102526. [PMID: 35344844 PMCID: PMC9167772 DOI: 10.1016/j.conb.2022.102526] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/11/2022] [Accepted: 02/15/2022] [Indexed: 12/25/2022]
Abstract
While aggression is often conceptualized as a highly stereotyped, innate behavior, individuals within a species exhibit a surprising amount of variability in the frequency, intensity, and targets of their aggression. While differences in genetics are a source of some of this variation across individuals (estimates place the heritability of behavior at around 25-30%), a critical driver of variability is previous life experience. A wide variety of social experiences, including sexual, parental, and housing experiences can facilitate "persistent" aggressive states, suggesting that these experiences engage a common set of synaptic and molecular mechanisms that act on dedicated neural circuits for aggression. It has long been known that sex steroid hormones are powerful modulators of behavior, and also, that levels of these hormones are themselves modulated by experience. Several recent studies have started to unravel how experience-dependent hormonal changes during adulthood can create a cascade of molecular, synaptic, and circuit changes that enable behavioral persistence through circuit level remodeling. Here, we propose that sex steroid hormones facilitate persistent aggressive states by changing the relationship between neural activity and an aggression "threshold".
Collapse
|
30
|
Liu M, Kim DW, Zeng H, Anderson DJ. Make war not love: The neural substrate underlying a state-dependent switch in female social behavior. Neuron 2022; 110:841-856.e6. [PMID: 34982958 PMCID: PMC8897222 DOI: 10.1016/j.neuron.2021.12.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 01/20/2023]
Abstract
Female mice exhibit opposing social behaviors toward males depending on their reproductive state: virgins display sexual receptivity (lordosis behavior), while lactating mothers attack. How a change in reproductive state produces a qualitative switch in behavioral response to the same conspecific stimulus is unknown. Using single-cell RNA-seq, we identify two distinct subtypes of estrogen receptor-1-positive neurons in the ventrolateral subdivision of the female ventromedial hypothalamus (VMHvl) and demonstrate that they causally control sexual receptivity and aggressiveness in virgins and lactating mothers, respectively. Between- and within-subject bulk-calcium recordings from each subtype reveal that aggression-specific cells acquire an increased responsiveness to social cues during the transition from virginity to maternity, while the responsiveness of the mating-specific population appears unchanged. These results demonstrate that reproductive-state-dependent changes in the relative activity of transcriptomically distinct neural subtypes can underlie categorical switches in behavior associated with physiological state changes.
Collapse
Affiliation(s)
- Mengyu Liu
- Program in Neurobiology, California Institute of Technology, Pasadena, CA, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA; Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dong-Wook Kim
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA; Allen Institute for Brain Science, Seattle WA 98109, USA
| | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle WA 98109, USA
| | - David J Anderson
- Program in Neurobiology, California Institute of Technology, Pasadena, CA, USA; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA; Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
31
|
Fulenwider HD, Caruso MA, Ryabinin AE. Manifestations of domination: Assessments of social dominance in rodents. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12731. [PMID: 33769667 PMCID: PMC8464621 DOI: 10.1111/gbb.12731] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/31/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023]
Abstract
Social hierarchies are ubiquitous features of virtually all animal groups. The varying social ranks of members within these groups have profound effects on both physical and emotional health, with lower-ranked individuals typically being the most adversely affected by their respective ranks. Thus, reliable measures of social dominance in preclinical rodent models are necessary to better understand the effects of an individual's social rank on other behaviors and physiological processes. In this review, we outline the primary methodologies used to assess social dominance in various rodent species: those that are based on analyses of agonistic behaviors, and those that are based on resource competition. In synthesizing this review, we conclude that assays based on resource competition may be better suited to characterize social dominance in a wider variety of rodent species and strains, and in both males and females. Lastly, albeit expectedly, we demonstrate that similarly to many other areas of preclinical research, studies incorporating female subjects are lacking in comparison to those using males. These findings emphasize the need for an increased number of studies assessing social dominance in females to form a more comprehensive understanding of this behavioral phenomenon.
Collapse
Affiliation(s)
- Hannah D. Fulenwider
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | - Maya A. Caruso
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| | - Andrey E. Ryabinin
- Department of Behavioral NeuroscienceOregon Health & Science UniversityPortlandORUSA
| |
Collapse
|
32
|
Oliveira VEDM, Bakker J. Neuroendocrine regulation of female aggression. Front Endocrinol (Lausanne) 2022; 13:957114. [PMID: 36034455 PMCID: PMC9399833 DOI: 10.3389/fendo.2022.957114] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Classically the neurobiology of aggression has been studied exclusively in males. Thus, females have been considered mildly aggressive except during lactation. Interestingly, recent studies in rodents and humans have revealed that non-lactating females can show exacerbated and pathological aggression similarly to males. This review provides an overview of recent findings on the neuroendocrine mechanisms regulating aggressive behavior in females. In particular, the focus will be on novel rodent models of exaggerated aggression established in non-lactating females. Among the neuromodulatory systems influencing female aggression, special attention has been given to sex-steroids and sex-steroid-sensitive neuronal populations (i.e., the core nuclei of the neural pathway of aggression) as well as to the neuropeptides oxytocin and vasopressin which are major players in the regulation of social behaviors.
Collapse
|
33
|
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: 2.5] [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
|
34
|
Huang S, Li G, Pan Y, Liu J, Zhao J, Zhang X, Lu W, Wan X, Krebs CJ, Wang Z, Han W, Zhang Z. Population variation alters aggression-associated oxytocin and vasopressin expressions in brains of Brandt's voles in field conditions. Front Zool 2021; 18:56. [PMID: 34717666 PMCID: PMC8557550 DOI: 10.1186/s12983-021-00441-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 10/12/2021] [Indexed: 11/29/2022] Open
Abstract
Density-dependent change in aggressive behavior contributes to the population regulation of many small rodents, but the underlying neurological mechanisms have not been examined in field conditions. We hypothesized that crowding stress and aggression-associated oxytocin (OT) and arginine vasopressin (AVP) in specific regions of the brain may be closely related to aggressive behaviors and population changes of small rodents. We analyzed the association of OT and AVP expression, aggressive behavior, and population density of Brandt’s voles in 24 large semi-natural enclosures (0.48 ha each) in Inner Mongolia grassland. We tested the effects of population density on the OT/AVP system and aggressive behavior by experimentally manipulating populations of Brandt’s voles in the grassland enclosures. High density was positively and significantly associated with more aggressive behavior, and increased expression of mRNA and protein of AVP and its receptor, but decreased expression of mRNA and protein of OT and its receptor in specific brain regions of the voles. Our study suggests that changes in OT/AVP expression are likely a result of the increased psychosocial stress that these voles experience during overcrowding, and thus the OT/AVP system can be used as indicators of density-dependent stressors in Brandt’s voles.
Collapse
Affiliation(s)
- Shuli Huang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guoliang Li
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongliang Pan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,School of Medicine, Huzhou University, Huzhou, 313000, China
| | - Jing Liu
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jidong Zhao
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xin Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Lu
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinrong Wan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Charles J Krebs
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, 32306-1270, USA
| | - Wenxuan Han
- College of Resources and Environmental Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
35
|
Herrero MJ, Wang L, Hernandez-Pineda D, Banerjee P, Matos HY, Goodrich M, Panigrahi A, Smith NA, Corbin JG. Sex-Specific Social Behavior and Amygdala Proteomic Deficits in Foxp2 +/- Mutant Mice. Front Behav Neurosci 2021; 15:706079. [PMID: 34421555 PMCID: PMC8374433 DOI: 10.3389/fnbeh.2021.706079] [Citation(s) in RCA: 8] [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: 05/06/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022] Open
Abstract
In humans, mutations in the transcription factor encoding gene, FOXP2, are associated with language and Autism Spectrum Disorders (ASD), the latter characterized by deficits in social interactions. However, little is known regarding the function of Foxp2 in male or female social behavior. Our previous studies in mice revealed high expression of Foxp2 within the medial subnucleus of the amygdala (MeA), a limbic brain region highly implicated in innate social behaviors such as mating, aggression, and parental care. Here, using a comprehensive panel of behavioral tests in male and female Foxp2 +/- heterozygous mice, we investigated the role Foxp2 plays in MeA-linked innate social behaviors. We reveal significant deficits in olfactory processing, social interaction, mating, aggressive, and parental behaviors. Interestingly, some of these deficits are displayed in a sex-specific manner. To examine the consequences of Foxp2 loss of function specifically in the MeA, we conducted a proteomic analysis of microdissected MeA tissue. This analyses revealed putative sex differences expression of a host of proteins implicated in neuronal communication, connectivity, and dopamine signaling. Consistent with this, we discovered that MeA Foxp2-lineage cells were responsive to dopamine with differences between males and females. Thus, our findings reveal a central and sex-specific role for Foxp2 in social behavior and MeA function.
Collapse
Affiliation(s)
- Maria Jesus Herrero
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Li Wang
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Payal Banerjee
- Center for Genomic Medicine, Children’s National Hospital, Washington, DC, United States
| | - Heidi Y. Matos
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Meredith Goodrich
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Aswini Panigrahi
- Center for Cancer and Immunology Research, Children’s National Hospital, Washington, DC, United States
| | - Nathan Anthony Smith
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| | - Joshua G. Corbin
- Center for Neuroscience Research, Children’s National Hospital, Washington, DC, United States
| |
Collapse
|
36
|
Zheng DJ, Singh A, Phelps SM. Conservation and dimorphism in androgen receptor distribution in Alston's singing mouse (Scotinomys teguina). J Comp Neurol 2021; 529:2539-2557. [PMID: 33576501 DOI: 10.1002/cne.25108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/19/2022]
Abstract
Because of their roles in courtship and intrasexual competition, sexual displays are often sexually dimorphic, but we know little about the mechanisms that produce such dimorphism. Among mammals, one example is the vocalization of Alston's singing mouse (Scotinomys teguina), which consists of a series of rapidly repeated, frequency-modulated notes. The rate and duration of songs is sexually dimorphic and androgen responsive. To understand the neuronal mechanisms underlying this sexual dimorphism, we map the sites of androgen sensitivity throughout the brain, focusing analysis along a pathway that spans from limbic structures to vocal motor regions. We find widespread expression of AR immunoreactivity (AR-ir) throughout limbic structures important for social behavior and vocalization, including the lateral septum, extended amygdala, preoptic area and hypothalamus. We also find extensive AR staining along previously documented vocal motor pathways, including the periaqueductal gray, parabrachial nucleus, and nucleus ambiguus, the last of which innervates intrinsic laryngeal muscles. Lastly, AR-ir is also evident in sensory areas such as the medial geniculate, inferior, and superior colliculi. A quantitative analysis revealed that males exhibited more AR-ir than females, a pattern that was most pronounced in the hypothalamus. Despite the elaboration of vocalization in singing mice, comparison with prior literature suggests that the broad pattern of AR-ir may be conserved across a wide range of rodents. Together these data identify brain nuclei well positioned to shape the sexually dimorphic vocalization of S. teguina and suggest that such androgen modulation of vocalization is evolutionary conserved among rodents.
Collapse
Affiliation(s)
- Da-Jiang Zheng
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Aditi Singh
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Steven M Phelps
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
37
|
Oliveira VEDM, Lukas M, Wolf HN, Durante E, Lorenz A, Mayer AL, Bludau A, Bosch OJ, Grinevich V, Egger V, de Jong TR, Neumann ID. Oxytocin and vasopressin within the ventral and dorsal lateral septum modulate aggression in female rats. Nat Commun 2021; 12:2900. [PMID: 34006875 PMCID: PMC8131389 DOI: 10.1038/s41467-021-23064-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 03/09/2021] [Indexed: 02/03/2023] Open
Abstract
In contrast to male rats, aggression in virgin female rats has been rarely studied. Here, we established a rat model of enhanced aggression in females using a combination of social isolation and aggression-training to specifically investigate the involvement of the oxytocin (OXT) and arginine vasopressin (AVP) systems within the lateral septum (LS). Using neuropharmacological, optogenetic, chemogenetic as well as microdialysis approaches, we revealed that enhanced OXT release within the ventral LS (vLS), combined with reduced AVP release within the dorsal LS (dLS), is required for aggression in female rats. Accordingly, increased activity of putative OXT receptor-positive neurons in the vLS, and decreased activity of putative AVP receptor-positive neurons in the dLS, are likely to underly aggression in female rats. Finally, in vitro activation of OXT receptors in the vLS increased tonic GABAergic inhibition of dLS neurons. Overall, our data suggest a model showing that septal release of OXT and AVP differentially affects aggression in females by modulating the inhibitory tone within LS sub-networks.
Collapse
Affiliation(s)
- Vinícius Elias de Moura Oliveira
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Michael Lukas
- Department of Neurobiology and Animal Physiology, Neurophysiology, University of Regensburg, Regensburg, Germany
| | - Hannah Nora Wolf
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Elisa Durante
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Alexandra Lorenz
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Anna-Lena Mayer
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Anna Bludau
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Oliver J Bosch
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Veronica Egger
- Department of Neurobiology and Animal Physiology, Neurophysiology, University of Regensburg, Regensburg, Germany
| | - Trynke R de Jong
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany
- Medische Biobank Noord-Nederland B.V., Groningen, Netherlands
| | - Inga D Neumann
- Department of Neurobiology and Animal Physiology, Behavioural and Molecular Neurobiology, University of Regensburg, Universitaetstraße, Regensburg, Bavaria, Germany.
| |
Collapse
|
38
|
Zhu Z, Ma Q, Miao L, Yang H, Pan L, Li K, Zeng LH, Zhang X, Wu J, Hao S, Lin S, Ma X, Mai W, Feng X, Hao Y, Sun L, Duan S, Yu YQ. A substantia innominata-midbrain circuit controls a general aggressive response. Neuron 2021; 109:1540-1553.e9. [DOI: 10.1016/j.neuron.2021.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/08/2020] [Accepted: 02/26/2021] [Indexed: 02/06/2023]
|
39
|
Sabbi KH, Emery Thompson M, Machanda ZP, Otali E, Wrangham RW, Muller MN. Sex differences in early experience and the development of aggression in wild chimpanzees. Proc Natl Acad Sci U S A 2021; 118:e2017144118. [PMID: 33727418 PMCID: PMC8000022 DOI: 10.1073/pnas.2017144118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sex differences in physical aggression occur across human cultures and are thought to be influenced by active sex role reinforcement. However, sex differences in aggression also exist in our close evolutionary relatives, chimpanzees, who do not engage in active teaching, but do exhibit long juvenile periods and complex social systems that allow differential experience to shape behavior. Here we ask whether early life exposure to aggression is sexually dimorphic in wild chimpanzees and, if so, whether other aspects of early sociality contribute to this difference. Using 13 y of all-occurrence aggression data collected from the Kanyawara community of chimpanzees (2005 to 2017), we determined that young male chimpanzees were victims of aggression more often than females by between 4 and 5 (i.e., early in juvenility). Combining long-term aggression data with data from a targeted study of social development (2015 to 2017), we found that two potential risk factors for aggression-time spent near adult males and time spent away from mothers-did not differ between young males and females. Instead, the major risk factor for receiving aggression was the amount of aggression that young chimpanzees displayed, which was higher for males than females throughout the juvenile period. In multivariate models, sex did not mediate this relationship, suggesting that other chimpanzees did not target young males specifically, but instead responded to individual behavior that differed by sex. Thus, social experience differed by sex even in the absence of explicit gender socialization, but experiential differences were shaped by early-emerging sex differences in behavior.
Collapse
Affiliation(s)
- Kris H Sabbi
- Department of Anthropology, Tufts University, Medford, MA 02155;
- Department of Anthropology, University of New Mexico, Albuquerque, NM 87131
| | - Melissa Emery Thompson
- Department of Anthropology, University of New Mexico, Albuquerque, NM 87131
- Kibale Chimpanzee Project, Fort Portal, Uganda
| | - Zarin P Machanda
- Department of Anthropology, Tufts University, Medford, MA 02155
- Kibale Chimpanzee Project, Fort Portal, Uganda
| | - Emily Otali
- Kibale Chimpanzee Project, Fort Portal, Uganda
| | - Richard W Wrangham
- Kibale Chimpanzee Project, Fort Portal, Uganda
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 87131
| | - Martin N Muller
- Department of Anthropology, University of New Mexico, Albuquerque, NM 87131
- Kibale Chimpanzee Project, Fort Portal, Uganda
| |
Collapse
|
40
|
Wei D, Talwar V, Lin D. Neural circuits of social behaviors: Innate yet flexible. Neuron 2021; 109:1600-1620. [PMID: 33705708 DOI: 10.1016/j.neuron.2021.02.012] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/31/2020] [Accepted: 02/09/2021] [Indexed: 12/16/2022]
Abstract
Social behaviors, such as mating, fighting, and parenting, are fundamental for survival of any vertebrate species. All members of a species express social behaviors in a stereotypical and species-specific way without training because of developmentally hardwired neural circuits dedicated to these behaviors. Despite being innate, social behaviors are flexible. The readiness to interact with a social target or engage in specific social acts can vary widely based on reproductive state, social experience, and many other internal and external factors. Such high flexibility gives vertebrates the ability to release the relevant behavior at the right moment and toward the right target. This maximizes reproductive success while minimizing the cost and risk associated with behavioral expression. Decades of research have revealed the basic neural circuits underlying each innate social behavior. The neural mechanisms that support behavioral plasticity have also started to emerge. Here we provide an overview of these social behaviors and their underlying neural circuits and then discuss in detail recent findings regarding the neural processes that support the flexibility of innate social behaviors.
Collapse
Affiliation(s)
- Dongyu Wei
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Vaishali Talwar
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Dayu Lin
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA; Department of Psychiatry, New York University School of Medicine, New York, NY, USA; Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
41
|
Schretter CE, Aso Y, Robie AA, Dreher M, Dolan MJ, Chen N, Ito M, Yang T, Parekh R, Branson KM, Rubin GM. Cell types and neuronal circuitry underlying female aggression in Drosophila. eLife 2020; 9:58942. [PMID: 33141021 PMCID: PMC7787668 DOI: 10.7554/elife.58942] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
Aggressive social interactions are used to compete for limited resources and are regulated by complex sensory cues and the organism’s internal state. While both sexes exhibit aggression, its neuronal underpinnings are understudied in females. Here, we identify a population of sexually dimorphic aIPg neurons in the adult Drosophila melanogaster central brain whose optogenetic activation increased, and genetic inactivation reduced, female aggression. Analysis of GAL4 lines identified in an unbiased screen for increased female chasing behavior revealed the involvement of another sexually dimorphic neuron, pC1d, and implicated aIPg and pC1d neurons as core nodes regulating female aggression. Connectomic analysis demonstrated that aIPg neurons and pC1d are interconnected and suggest that aIPg neurons may exert part of their effect by gating the flow of visual information to descending neurons. Our work reveals important regulatory components of the neuronal circuitry that underlies female aggressive social interactions and provides tools for their manipulation.
Collapse
Affiliation(s)
| | - Yoshinori Aso
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Alice A Robie
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Marisa Dreher
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Michael-John Dolan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States.,Current address: Department of Neurology and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Nan Chen
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Masayoshi Ito
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Tansy Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Ruchi Parekh
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Kristin M Branson
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Gerald M Rubin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
42
|
Neural mechanisms of aggression across species. Nat Neurosci 2020; 23:1317-1328. [PMID: 33046890 DOI: 10.1038/s41593-020-00715-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 08/31/2020] [Indexed: 12/21/2022]
Abstract
Aggression is a social behavior essential for securing resources and defending oneself and family. Thanks to its indispensable function in competition and thus survival, aggression exists widely across animal species, including humans. Classical works from Tinbergen and Lorenz concluded that instinctive behaviors including aggression are mediated by hardwired brain circuitries that specialize in processing certain sensory inputs to trigger stereotyped motor outputs. They further suggest that instinctive behaviors are influenced by an animal's internal state and past experiences. Following this conceptual framework, here we review our current understanding regarding the neural substrates underlying aggression generation, highlighting an evolutionarily conserved 'core aggression circuit' composed of four subcortical regions. We further discuss the neural mechanisms that support changes in aggression based on the animal's internal state. We aim to provide an overview of features of aggression and the relevant neural substrates across species, highlighting findings in rodents, primates and songbirds.
Collapse
|
43
|
Goodwin NL, Nilsson SRO, Golden SA. Rage Against the Machine: Advancing the study of aggression ethology via machine learning. Psychopharmacology (Berl) 2020; 237:2569-2588. [PMID: 32647898 PMCID: PMC7502501 DOI: 10.1007/s00213-020-05577-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/01/2020] [Indexed: 12/24/2022]
Abstract
RATIONALE Aggression, comorbid with neuropsychiatric disorders, exhibits with diverse clinical presentations and places a significant burden on patients, caregivers, and society. This diversity is observed because aggression is a complex behavior that can be ethologically demarcated as either appetitive (rewarding) or reactive (defensive), each with its own behavioral characteristics, functionality, and neural basis that may transition from adaptive to maladaptive depending on genetic and environmental factors. There has been a recent surge in the development of preclinical animal models for studying appetitive aggression-related behaviors and identifying the neural mechanisms guiding their progression and expression. However, adoption of these procedures is often impeded by the arduous task of manually scoring complex social interactions. Manual observations are generally susceptible to observer drift, long analysis times, and poor inter-rater reliability, and are further incompatible with the sampling frequencies required of modern neuroscience methods. OBJECTIVES In this review, we discuss recent advances in the preclinical study of appetitive aggression in mice, paired with our perspective on the potential for machine learning techniques in producing automated, robust scoring of aggressive social behavior. We discuss critical considerations for implementing valid computer classifications within behavioral pharmacological studies. KEY RESULTS Open-source automated classification platforms can match or exceed the performance of human observers while removing the confounds of observer drift, bias, and inter-rater reliability. Furthermore, unsupervised approaches can identify previously uncharacterized aggression-related behavioral repertoires in model species. DISCUSSION AND CONCLUSIONS Advances in open-source computational approaches hold promise for overcoming current manual annotation caveats while also introducing and generalizing computational neuroethology to the greater behavioral neuroscience community. We propose that currently available open-source approaches are sufficient for overcoming the main limitations preventing wide adoption of machine learning within the context of preclinical aggression behavioral research.
Collapse
Affiliation(s)
- Nastacia L Goodwin
- Department of Biological Structure, University of Washington, Seattle, WA, USA
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| | - Simon R O Nilsson
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Sam A Golden
- Department of Biological Structure, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
- Center of Excellence in Neurobiology of Addiction, Pain, and Emotion (NAPE), University of Washington, Seattle, WA, USA.
| |
Collapse
|
44
|
Huang S, Li G, Pan Y, Song M, Zhao J, Wan X, Krebs CJ, Wang Z, Han W, Zhang Z. Density-induced social stress alters oxytocin and vasopressin activities in the brain of a small rodent species. Integr Zool 2020; 16:149-159. [PMID: 32652776 PMCID: PMC7891312 DOI: 10.1111/1749-4877.12467] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
It is known that social stress could alter oxytocin (OT) and arginine‐vasopressin (AVP) expression in specific regions of brains which regulate the aggressive behavior of small rodents, but the effects of density‐induced social stress are still unknown. Brandt's voles (Lasiopodomys brandtii) are small herbivores in the grassland of China, but the underlying neurological mechanism of population regulation is still unknown. We tested the effects of housing density of Brandt's voles on OT/AVP system with physical contact (allowing aggression) and without physical contact (not allowing aggression) under laboratory conditions. Then, we tested the effects of paired‐aggression (no density effect) of Brandt's voles on OT/AVP system under laboratory conditions. We hypothesized that high density would increase aggression among animals which would then increase AVP but reduce OT in brains of animals. Our results showed that high housing density induced more aggressive behavior. We found high‐density‐induced social stress (with or without physical contact) and direct aggression significantly increased expression of mRNA and protein of AVP and its receptor, but decreased expression of mRNA and protein of OT and its receptor in specific brain regions of voles. The results suggest that density‐dependent change of OT/AVP systems may play a significant role in the population regulation of small rodents by altering density‐dependent aggressive behavior.
Collapse
Affiliation(s)
- Shuli Huang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Guoliang Li
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Yongliang Pan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,School of Medicine, Huzhou University, Huzhou, China
| | - Mingjing Song
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Science, Beijing, China
| | - Jidong Zhao
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| | - Xinrong Wan
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Charles J Krebs
- Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, Florida, USA
| | - Wenxuan Han
- College of Resources and Environmental Sciences, China Agricultural University, Beijing, China
| | - Zhibin Zhang
- State Key Laboratory of Integrated Pest Management, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
45
|
Sex Differences in Biophysical Signatures across Molecularly Defined Medial Amygdala Neuronal Subpopulations. eNeuro 2020; 7:ENEURO.0035-20.2020. [PMID: 32493755 PMCID: PMC7333980 DOI: 10.1523/eneuro.0035-20.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/20/2020] [Indexed: 12/29/2022] Open
Abstract
The medial amygdala (MeA) is essential for processing innate social and non-social behaviors, such as territorial aggression and mating, which display in a sex-specific manner. While sex differences in cell numbers and neuronal morphology in the MeA are well established, if and how these differences extend to the biophysical level remain unknown. Our previous studies revealed that expression of the transcription factors, Dbx1 and Foxp2, during embryogenesis defines separate progenitor pools destined to generate different subclasses of MEA inhibitory output neurons. We have also previously shown that Dbx1-lineage and Foxp2-lineage neurons display different responses to innate olfactory cues and in a sex-specific manner. To examine whether these neurons also possess sex-specific biophysical signatures, we conducted a multidimensional analysis of the intrinsic electrophysiological profiles of these transcription factor defined neurons in the male and female MeA. We observed striking differences in the action potential (AP) spiking patterns across lineages, and across sex within each lineage, properties known to be modified by different voltage-gated ion channels. To identify the potential mechanism underlying the observed lineage-specific and sex-specific differences in spiking adaptation, we conducted a phase plot analysis to narrow down putative ion channel candidates. Of these candidates, we found a subset expressed in a lineage-biased and/or sex-biased manner. Thus, our results uncover neuronal subpopulation and sex differences in the biophysical signatures of developmentally defined MeA output neurons, providing a potential physiological substrate for how the male and female MeA may process social and non-social cues that trigger innate behavioral responses.
Collapse
|
46
|
Wohl M, Ishii K, Asahina K. Layered roles of fruitless isoforms in specification and function of male aggression-promoting neurons in Drosophila. eLife 2020; 9:e52702. [PMID: 32314957 PMCID: PMC7173971 DOI: 10.7554/elife.52702] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 04/03/2020] [Indexed: 12/20/2022] Open
Abstract
Inter-male aggressive behavior is a prominent sexually dimorphic behavior. Neural circuits that underlie aggressive behavior are therefore likely under the control of sex-determining genes. However, the neurogenetic mechanism that generates sex-specific aggressive behavior remains largely unknown. Here, we found that a neuronal class specified by one of the Drosophila sex determining genes, fruitless (fru), belongs to the neural circuit that generates male-type aggressive behavior. This neuronal class can promote aggressive behavior independent of another sex determining gene, doublesex (dsx), although dsx is involved in ensuring that aggressive behavior is performed only toward males. We also found that three fru isoforms with different DNA binding domains show a division of labor on male aggressive behaviors. A dominant role of fru in specifying sex-specific aggressive behavior may underscore a genetic mechanism that allows male-type aggressive behavior to evolve at least partially independently from courtship behavior, which is under different selective pressures.
Collapse
Affiliation(s)
- Margot Wohl
- Molecular Neurobiology Laboratory, The Salk Institute for Biological StudiesLa JollaUnited States
- Neuroscience Graduate Program, University of CaliforniaSan DiegoUnited States
| | - Kenichi Ishii
- Molecular Neurobiology Laboratory, The Salk Institute for Biological StudiesLa JollaUnited States
- Neuroscience Graduate Program, University of CaliforniaSan DiegoUnited States
| | - Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological StudiesLa JollaUnited States
| |
Collapse
|
47
|
Gegenhuber B, Tollkuhn J. Signatures of sex: Sex differences in gene expression in the vertebrate brain. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e348. [PMID: 31106965 PMCID: PMC6864223 DOI: 10.1002/wdev.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | |
Collapse
|
48
|
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: 3.5] [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
|
49
|
Newman EL, Covington HE, Suh J, Bicakci MB, Ressler KJ, DeBold JF, Miczek KA. Fighting Females: Neural and Behavioral Consequences of Social Defeat Stress in Female Mice. Biol Psychiatry 2019; 86:657-668. [PMID: 31255250 PMCID: PMC6788975 DOI: 10.1016/j.biopsych.2019.05.005] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND Despite the twofold higher prevalence of major depressive and posttraumatic stress disorders in women compared with men, most clinical and preclinical studies have focused on male subjects. We used an ethological murine model to study several cardinal symptoms of affective disorders in the female targets of female aggression. METHODS Intact Swiss Webster (CFW) female resident mice were housed with castrated male mice and tested for aggression toward female intruders. For 10 days, aggressive CFW female residents defeated C57BL/6J (B6) female intruders during 5-minute encounters. Measures of corticosterone, c-Fos activation in hypothalamic and limbic structures, and species-typical behaviors were collected from defeated and control females. Ketamine (20 mg/kg) was tested for its potential to reverse stress-induced social deficits. RESULTS Housed with a castrated male mouse, most intact resident CFW females readily attacked unfamiliar B6 female intruders, inflicting >40 bites in a 5-minute encounter. Compared with controls, defeated B6 females exhibited elevated plasma corticosterone and increased c-Fos activation in the medial amygdala, ventral lateral septum, ventromedial hypothalamus, and hypothalamic paraventricular nucleus. Chronically defeated females also showed vigilance-like behavior and deficits in social interactions, novel object investigation, and nesting. The duration of social interactions increased 24 hours after chronically defeated female mice received a systemic dose of ketamine. CONCLUSIONS These findings demonstrate that CFW female mice living with male conspecifics can be used as aggressive residents in an ethological model of female social defeat stress. These novel behavioral methods will encourage further studies of sex-specific neural, physiological, and behavioral adaptations to chronic stress and the biological bases for interfemale aggression.
Collapse
Affiliation(s)
- Emily L Newman
- Psychology Department, Tufts University, Medford, Massachusetts
| | | | - Junghyup Suh
- Division of Depression and Anxiety Disorders and Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | | | - Kerry J Ressler
- Division of Depression and Anxiety Disorders and Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, Massachusetts
| | - Joseph F DeBold
- Psychology Department, Tufts University, Medford, Massachusetts
| | - Klaus A Miczek
- Psychology Department, Tufts University, Medford, Massachusetts; Department of Neuroscience, Tufts University, Boston, Massachusetts.
| |
Collapse
|
50
|
Hahn JD, Fink G, Kruk MR, Stanley BG. Editorial: Current Views of Hypothalamic Contributions to the Control of Motivated Behaviors. Front Syst Neurosci 2019; 13:32. [PMID: 31456668 PMCID: PMC6700385 DOI: 10.3389/fnsys.2019.00032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/08/2019] [Indexed: 11/25/2022] Open
Affiliation(s)
- Joel D Hahn
- Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| | - George Fink
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Menno R Kruk
- Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, Netherlands
| | - B Glenn Stanley
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|