1
|
Han C, Cheung VCK, Chan RHM. Aging amplifies sex differences in low alpha and low beta EEG oscillations. Neuroimage 2025; 312:121231. [PMID: 40252876 DOI: 10.1016/j.neuroimage.2025.121231] [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: 12/29/2024] [Revised: 03/30/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025] Open
Abstract
Biological sex profoundly shapes brain function, yet its precise influence on neural oscillations was poorly understood. Despite decades of research, studies investigating sex-based variations in electroencephalographic (EEG) signals have yielded inconsistent findings that obstructs what may be a potentially crucial source of inter-individual variability in brain function. To address this, we analyzed five publicly available resting-state datasets, comprising EEG data (n = 445) and iEEG data (n = 103). Three age ranges were defined, young adult (YA, 18-30 years), middle-aged adult (MA, 30-55 years) and older adult (OA, 55-80 years). Our results revealed striking age-dependent sex differences: OA group exhibited robust sex differences, with males showing heightened low alpha (8-9 Hz) activity in temporal regions and attenuated low beta (16-20 Hz) oscillations in parietal-occipital areas compared to females. Intriguingly, these sex-specific patterns were absent in YA group, suggesting a complex interplay between sex and aging in shaping brain dynamics. The MA groups fall in between YA and OA group. The increase of low beta band activity in older female adults is strongly associated with hip size and BMI. Furthermore, we identified consistent sex-related activity in the precentral gyrus with the results of scalp EEG, potentially driving the observed scalp EEG differences. This multi-level analysis allowed us to bridge the gap between cortical and scalp-level observations, providing a more comprehensive picture of sex-related neural dynamics. The distinct associations between sex-specific oscillatory patterns and several lifestyle factors demonstrates the complex interplay between sex, age, and neural oscillations, revealing the variability in brain dynamics. Our findings highlight the importance of careful demographic consideration in EEG research design to ensure fairness in capturing the full spectrum of neurophysiological diversity.
Collapse
Affiliation(s)
- Chuanliang Han
- School of Biomedical Sciences and The Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Electrical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Vincent C K Cheung
- School of Biomedical Sciences and The Gerald Choa Neuroscience Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Rosa H M Chan
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
2
|
Kaplan HS, Horvath PM, Rahman MM, Dulac C. The neurobiology of parenting and infant-evoked aggression. Physiol Rev 2025; 105:315-381. [PMID: 39146250 DOI: 10.1152/physrev.00036.2023] [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: 09/21/2023] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Parenting behavior comprises a variety of adult-infant and adult-adult interactions across multiple timescales. The state transition from nonparent to parent requires an extensive reorganization of individual priorities and physiology and is facilitated by combinatorial hormone action on specific cell types that are integrated throughout interconnected and brainwide neuronal circuits. In this review, we take a comprehensive approach to integrate historical and current literature on each of these topics across multiple species, with a focus on rodents. New and emerging molecular, circuit-based, and computational technologies have recently been used to address outstanding gaps in our current framework of knowledge on infant-directed behavior. This work is raising fundamental questions about the interplay between instinctive and learned components of parenting and the mutual regulation of affiliative versus agonistic infant-directed behaviors in health and disease. Whenever possible, we point to how these technologies have helped gain novel insights and opened new avenues of research into the neurobiology of parenting. We hope this review will serve as an introduction for those new to the field, a comprehensive resource for those already studying parenting, and a guidepost for designing future studies.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Patricia M Horvath
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Mohammed Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| |
Collapse
|
3
|
Liao CP, Majeed M, Hobert O. Experience-dependent, sexually dimorphic synaptic connectivity defined by sex-specific cadherin expression. SCIENCE ADVANCES 2024; 10:eadq9183. [PMID: 39536115 PMCID: PMC11559607 DOI: 10.1126/sciadv.adq9183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Early-life experience influences subsequent maturation and function of the adult brain, sometimes even in a sex-specific manner, but underlying molecular mechanisms are poorly understood. We describe here how juvenile experience defines sexually dimorphic synaptic connectivity in the adult Caenorhabditis elegans nervous system. Starvation of juvenile males disrupts serotonin-dependent activation of the CREB transcription factor in a nociceptive sensory neuron, PHB. CREB acts through a cascade of transcription factors to control expression of an atypical cadherin protein, FMI-1/Flamingo/CELSR. During postembryonic development, FMI-1 promotes and maintains synaptic connectivity of PHB to a command interneuron, AVA, in both sexes, but a serotonin-dependent transcriptional regulatory cassette antagonizes FMI-1 expression in males, thereby establishing sexually dimorphic connectivity between PHB and AVA. A critical regulatory node is the CREB-target LIN-29, a Zn finger transcription factor that integrates four layers of information: sexual specificity, past experience, time and cell-type specificity. Our findings provide the mechanistic details of how an early juvenile experience defines sexually dimorphic synaptic connectivity.
Collapse
Affiliation(s)
- Chien-Po Liao
- Department of Biological Sciences, Columbia University, Howard Hughes Medical Institute, New York, NY 10027, USA
| | | | - Oliver Hobert
- Department of Biological Sciences, Columbia University, Howard Hughes Medical Institute, New York, NY 10027, USA
| |
Collapse
|
4
|
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
|
5
|
Liao CP, Majeed M, Hobert O. Experience-dependent, sexually dimorphic synaptic connectivity defined by sex-specific cadherin expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593207. [PMID: 38766005 PMCID: PMC11100761 DOI: 10.1101/2024.05.08.593207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
We describe here the molecular mechanisms by which juvenile experience defines patterns of sexually dimorphic synaptic connectivity in the adult nervous system of the nematode C. elegans. We show that starvation of juvenile males disrupts serotonin-dependent activation of the CREB transcription factor in a nociceptive sensory neuron, PHB. CREB acts through a cascade of transcription factors to control expression of an atypical cadherin protein, FMI-1/Flamingo. During postembryonic development, FMI-1/Flamingo has the capacity to promote and maintain synaptic connectivity of the PHB nociceptive sensory to a command interneuron, AVA, in both sexes, but the serotonin transcriptional regulatory cassette antagonizes FMI-1/Flamingo expression in males, thereby establishing sexually dimorphic connectivity between PHB and AVA. A critical regulatory node in this process is the CREB-target LIN-29, a Zn finger transcription factor which integrates four different layers of information - sexual specificity, past feeding status, time and cell-type specificity. Our findings provide the mechanistic details of how an early juvenile experience defines sexually dimorphic synaptic connectivity.
Collapse
Affiliation(s)
- Chien-Po Liao
- Department of Biological Sciences, Columbia University, Howard
Hughes Medical Institute, New York, NY 10027, USA
| | - Maryam Majeed
- Department of Biological Sciences, Columbia University, Howard
Hughes Medical Institute, New York, NY 10027, USA
- Present address: Allen Institute for Brain Science, Seattle,
USA
| | - Oliver Hobert
- Department of Biological Sciences, Columbia University, Howard
Hughes Medical Institute, New York, NY 10027, USA
| |
Collapse
|
6
|
Fang S, Luo Z, Wei Z, Qin Y, Zheng J, Zhang H, Jin J, Li J, Miao C, Yang S, Li Y, Liang Z, Yu XD, Zhang XM, Xiong W, Zhu H, Gan WB, Huang L, Li B. Sexually dimorphic control of affective state processing and empathic behaviors. Neuron 2024; 112:1498-1517.e8. [PMID: 38430912 DOI: 10.1016/j.neuron.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 03/05/2024]
Abstract
Recognizing the affective states of social counterparts and responding appropriately fosters successful social interactions. However, little is known about how the affective states are expressed and perceived and how they influence social decisions. Here, we show that male and female mice emit distinct olfactory cues after experiencing distress. These cues activate distinct neural circuits in the piriform cortex (PiC) and evoke sexually dimorphic empathic behaviors in observers. Specifically, the PiC → PrL pathway is activated in female observers, inducing a social preference for the distressed counterpart. Conversely, the PiC → MeA pathway is activated in male observers, evoking excessive self-grooming behaviors. These pathways originate from non-overlapping PiC neuron populations with distinct gene expression signatures regulated by transcription factors and sex hormones. Our study unveils how internal states of social counterparts are processed through sexually dimorphic mechanisms at the molecular, cellular, and circuit levels and offers insights into the neural mechanisms underpinning sex differences in higher brain functions.
Collapse
Affiliation(s)
- Shunchang Fang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhengyi Luo
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zicheng Wei
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuxin Qin
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jieyan Zheng
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hongyang Zhang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jianhua Jin
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jiali Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Chenjian Miao
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Shana Yang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yonglin Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zirui Liang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao-Dan Yu
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiao Min Zhang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wei Xiong
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Hongying Zhu
- Institute on Aging, Hefei, China and Brain Disorders, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | | | - Lianyan Huang
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou 510655, China.
| | - Boxing Li
- Neuroscience Program, Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China; Advanced Medical Technology Center, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou 510655, China.
| |
Collapse
|
7
|
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
|
8
|
Liu X, Feng X, Huang H, Huang K, Xu Y, Ye S, Tseng YT, Wei P, Wang L, Wang F. Male and female mice display consistent lifelong ability to address potential life-threatening cues using different post-threat coping strategies. BMC Biol 2022; 20:281. [PMID: 36522765 PMCID: PMC9753375 DOI: 10.1186/s12915-022-01486-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sex differences ranging from physiological functions to pathological disorders are developmentally hard-wired in a broad range of animals, from invertebrates to humans. These differences ensure that animals can display appropriate behaviors under a variety of circumstances, such as aggression, hunting, sleep, mating, and parental care, which are often thought to be important in the acquisition of resources, including territory, food, and mates. Although there are reports of an absence of sexual dimorphism in the context of innate fear, the question of whether there is sexual dimorphism of innate defensive behavior is still an open question. Therefore, an in-depth investigation to determine whether there are sex differences in developmentally hard-wired innate defensive behaviors in life-threatening circumstances is warranted. RESULTS We found that innate defensive behavioral responses to potentially life-threatening stimuli between males and females were indistinguishable over their lifespan. However, by using 3 dimensional (3D)-motion learning framework analysis, we found that males and females showed different behavioral patterns after escaping to the refuge. Specifically, the defensive "freezing" occurred primarily in males, whereas females were more likely to return directly to exploration. Moreover, there were also no estrous phase differences in innate defensive behavioral responses after looming stimuli. CONCLUSIONS Our results demonstrate that visually-evoked innate fear behavior is highly conserved throughout the lifespan in both males and females, while specific post-threat coping strategies depend on sex. These findings indicate that innate fear behavior is essential to both sexes and as such, there are no evolutionary-driven sex differences in defensive ability.
Collapse
Affiliation(s)
- Xue Liu
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaolong Feng
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Hongren Huang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kang Huang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Yang Xu
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuwei Ye
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Yu-Ting Tseng
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Pengfei Wei
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Liping Wang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China
| | - Feng Wang
- Shenzhen Key Lab of Translational Research for Brain Diseases, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
| |
Collapse
|
9
|
Westrick SE, Moss JB, Fischer EK. Who cares? An integrative approach to understanding the evolution of behavioural plasticity in parental care. Anim Behav 2022. [DOI: 10.1016/j.anbehav.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
10
|
Pinacho-Guendulain B, Montiel-Castro AJ, Ramos-Fernández G, Pacheco-López G. Social complexity as a driving force of gut microbiota exchange among conspecific hosts in non-human primates. Front Integr Neurosci 2022; 16:876849. [PMID: 36110388 PMCID: PMC9468716 DOI: 10.3389/fnint.2022.876849] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The emergent concept of the social microbiome implies a view of a highly connected biological world, in which microbial interchange across organisms may be influenced by social and ecological connections occurring at different levels of biological organization. We explore this idea reviewing evidence of whether increasing social complexity in primate societies is associated with both higher diversity and greater similarity in the composition of the gut microbiota. By proposing a series of predictions regarding such relationship, we evaluate the existence of a link between gut microbiota and primate social behavior. Overall, we find that enough empirical evidence already supports these predictions. Nonetheless, we conclude that studies with the necessary, sufficient, explicit, and available evidence are still scarce. Therefore, we reflect on the benefit of founding future analyses on the utility of social complexity as a theoretical framework.
Collapse
Affiliation(s)
- Braulio Pinacho-Guendulain
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Ciudad de México, Mexico
| | - Augusto Jacobo Montiel-Castro
- Department of Health Sciences, Metropolitan Autonomous University (UAM), Lerma, Mexico
- *Correspondence: Augusto Jacobo Montiel-Castro,
| | - Gabriel Ramos-Fernández
- Institute for Research on Applied Mathematics and Systems (IIMAS), National Autonomous University of Mexico (UNAM), Mexico City, Mexico
- Center for Complexity Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Gustavo Pacheco-López
- Department of Health Sciences, Metropolitan Autonomous University (UAM), Lerma, Mexico
- Gustavo Pacheco-López,
| |
Collapse
|
11
|
Kopachev N, Netser S, Wagner S. Sex-dependent features of social behavior differ between distinct laboratory mouse strains and their mixed offspring. iScience 2022; 25:103735. [PMID: 35098101 PMCID: PMC8783130 DOI: 10.1016/j.isci.2022.103735] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/25/2021] [Accepted: 12/31/2021] [Indexed: 11/26/2022] Open
Abstract
The survival of individuals of gregarious species depends on their social interactions. In humans, atypical social behavior is a hallmark of several psychopathological conditions, many of which have sex-specific manifestations. Various laboratory mouse strains are used to reveal the mechanisms mediating typical and atypical social behavior in mammals. Here, we used three social discrimination tests to characterize social behavior in males and females of three widely used laboratory mouse strains (C57BL/6J, BALB/c, and ICR). We found marked sex- and strain-specific differences in the behavior exhibited by subjects, in a test-dependent manner. Interestingly, some characteristics were strain-dependent, while others were sex-dependent. We then crossbred C57BL/6J and BALB/c mice and found that offspring of such crossbreeding exhibit social behavior which differs from both parental strains and depends on the specific combination of parental strains. Thus, social behavior of laboratory mice is sex- and strain-specific and depends on both genetic and environmental factors. Social investigation behavior of laboratory mice is highly strain- and sex-specific Some behavioral aspects are either strain- or sex-specific, but not both Mixed offspring of distinct strains behave differently from both parental strains The behavior of mixed offspring depends on the specific combination of parents
Collapse
|
12
|
Berrosteguieta I, Rosillo JC, Herrera ML, Olivera-Bravo S, Casanova G, Herranz-Pérez V, García-Verdugo JM, Fernández AS. Plasticity of cell proliferation in the retina of Austrolebias charrua fish under light and darkness conditions. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 3:100042. [DOI: 10.1016/j.crneur.2022.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022] Open
|
13
|
Johnson CS, Hong W, Micevych PE. Posterodorsal Medial Amygdala Regulation of Female Social Behavior: GABA versus Glutamate Projections. J Neurosci 2021; 41:8790-8800. [PMID: 34470806 PMCID: PMC8528505 DOI: 10.1523/jneurosci.1103-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 11/21/2022] Open
Abstract
Social behaviors, including reproductive behaviors, often display sexual dimorphism. Lordosis, the measure of female sexual receptivity, is one of the most apparent sexually dimorphic reproductive behaviors. Lordosis is regulated by estrogen and progesterone (P4) acting within a hypothalamic-limbic circuit, consisting of the arcuate, medial preoptic, and ventromedial nuclei of the hypothalamus. Social cues are integrated into the circuit through the amygdala. The posterodorsal part of the medial amygdala (MeApd) is involved in sexually dimorphic social and reproductive behaviors, and sends projections to hypothalamic neuroendocrine regions. GABA from the MeApd appears to facilitate social behaviors, while glutamate may play the opposite role. To test these hypotheses, adult female vesicular GABA transporter (VGAT)-Cre and vesicular glutamate transporter 2 (VGluT2)-Cre mice were transfected with halorhodopsin (eNpHR)-expressing or channelrhodopsin-expressing adeno-associated viruses (AAVs), respectively, in the MeApd. The lordosis quotient (LQ) was measured following either photoinhibition of VGAT or photoexcitation of VGluT2 neurons, and brains were assessed for c-Fos immunohistochemistry (IHC). Photoinhibition of VGAT neurons in the MeApd decreased LQ, and decreased c-Fos expression within VGAT neurons, within the MeApd as a whole, and within the ventrolateral part of the ventromedial nucleus (VMHvl). Photoexcitation of VGluT2 neurons did not affect LQ, but did increase time spent self-grooming, and increased c-Fos expression within VGluT2 neurons in the MeApd. Neither condition altered c-Fos expression in the medial preoptic nucleus (MPN) or the arcuate nucleus (ARH). These data support a role for MeApd GABA in the facilitation of lordosis. Glutamate from the MeApd does not appear to be directly involved in the lordosis circuit, but appears to direct behavior away from social interactions.SIGNIFICANCE STATEMENT Lordosis, the measure of female sexual receptivity, is a sexually dimorphic behavior regulated within a hypothalamic-limbic circuit. Social cues are integrated through the amygdala, and the posterodorsal part of the medial amygdala (MeApd) is involved in sexually dimorphic social and reproductive behaviors. Photoinhibition of GABAergic neurons in the MeApd inhibited lordosis, while photoactivation of glutamate neurons had no effect on lordosis, but increased self-grooming. These data support a role for MeApd GABA in the facilitation of social behaviors and MeApd glutamate projections in anti-social interactions.
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095
| | - Weizhe Hong
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Paul E Micevych
- Department of Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California 90095
- Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
14
|
Zilkha N, Sofer Y, Kashash Y, Kimchi T. The social network: Neural control of sex differences in reproductive behaviors, motivation, and response to social isolation. Curr Opin Neurobiol 2021; 68:137-151. [PMID: 33910083 PMCID: PMC8528716 DOI: 10.1016/j.conb.2021.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 12/30/2022]
Abstract
Social animal species present a vast repertoire of social interactions when encountering conspecifics. Reproduction-related behaviors, such as mating, parental care, and aggression, are some of the most rewarding types of social interactions and are also the most sexually dimorphic ones. This review focuses on rodent species and summarizes recent advances in neuroscience research that link sexually dimorphic reproductive behaviors to sexual dimorphism in their underlying neuronal circuits. Specifically, we present a few possible mechanisms governing sexually-dimorphic behaviors, by hypothalamic and reward-related brain regions. Sex differences in the neural response to social isolation in adulthood are also discussed, as well as future directions for comparative studies with naturally solitary species.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yizhak Sofer
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yael Kashash
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
15
|
Gilbert SF. Systemic racism, systemic sexism, and the embryological enterprise. Dev Biol 2021; 473:97-104. [PMID: 33609565 DOI: 10.1016/j.ydbio.2021.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 11/17/2022]
Abstract
The core of systemic racism and sexism is not merely an emphasis about human differences and thinking that another group of people is inferior to one's own. Rather, the institutional nature of racism or sexism establishes a permanent group hierarchy that is believed to reflect the laws of nature or the decrees of God. It thus becomes the norm of a culture to think and behave according to these rules. Notions of hierarchy became solidified into the Great Chain of Being during the Middle Ages, as did views concerning hereditary racial and gender superiority. During the Enlightenment, such classifications became established by philosophy and science. Starting in the 1800s, embryology and anthropology were used to provide evidence for the unilinear progression of species and races. The first evolutionary schemes were not "branching trees." In these schemes, women and non-white races were seen as embryonic or juvenile forms of the adult white male, and they were often depicted as intermediaries between the fully human and the animals. Such linear schemes of evolution remain part of popular culture and even some science, promoting the racism and sexism associated with them.
Collapse
Affiliation(s)
- Scott F Gilbert
- Department of Biology, Swarthmore College, Swarthmore, PA 19081, USA; Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
16
|
Chiu H, Hoopfer ED, Coughlan ML, Pavlou HJ, Goodwin SF, Anderson DJ. A circuit logic for sexually shared and dimorphic aggressive behaviors in Drosophila. Cell 2021; 184:507-520.e16. [PMID: 33382967 PMCID: PMC7856078 DOI: 10.1016/j.cell.2020.11.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/27/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022]
Abstract
Aggression involves both sexually monomorphic and dimorphic actions. How the brain implements these two types of actions is poorly understood. We have identified three cell types that regulate aggression in Drosophila: one type is sexually shared, and the other two are sex specific. Shared common aggression-promoting (CAP) neurons mediate aggressive approach in both sexes, whereas functionally downstream dimorphic but homologous cell types, called male-specific aggression-promoting (MAP) neurons in males and fpC1 in females, control dimorphic attack. These symmetric circuits underlie the divergence of male and female aggressive behaviors, from their monomorphic appetitive/motivational to their dimorphic consummatory phases. The strength of the monomorphic → dimorphic functional connection is increased by social isolation in both sexes, suggesting that it may be a locus for isolation-dependent enhancement of aggression. Together, these findings reveal a circuit logic for the neural control of behaviors that include both sexually monomorphic and dimorphic actions, which may generalize to other organisms.
Collapse
Affiliation(s)
- Hui Chiu
- Division of Biology and Biological Engineering 156-29, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA 91125, USA.
| | - Eric D Hoopfer
- Carleton College, 1 N. College St., Northfield, MN 55057, USA
| | - Maeve L Coughlan
- Mount Holyoke College, 50 College St., South Hadley, MA 01075, USA
| | - Hania J Pavlou
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - Stephen F Goodwin
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - David J Anderson
- Division of Biology and Biological Engineering 156-29, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA 91125, USA; Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
17
|
Zhang L, Li P, Ou H, Liu Z, Sun Y, Song Q, Hou L, Huang S, Meng Z, Liang L. Gender roles and behavioral problems in children with 21-hydroxylase deficiency in Southern China. Steroids 2021; 165:108754. [PMID: 33172829 DOI: 10.1016/j.steroids.2020.108754] [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: 04/16/2020] [Revised: 10/06/2020] [Accepted: 10/17/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE To assess the gender roles and behavioral outcomes in children with 21-hydroxylase deficiency (21-OHD) in Southern China. METHODS A total of 50 individuals with 21-OHD participated in our study, (30 boys and 20 girls), as well as another 19 age-matched non-affected relatives of patients (12 boys and 7 girls). Psychological adjustment was assessed with a preschool activity survey and a Conner parent symptom questionnaire was modified for retrospective reporting. RESULTS The response rate of the questionnaire in the control group was only 36.5%. All the patients were diagnosed with salt-wasting of 21-OHD. Our study revealed that the masculine score was higher in male patients with 21-OHD than male controls and female patients. Compared with that in the female 21-OHD patient group, the masculine score in the female control group was lower, while comparative masculinization was found in the male controls. Regarding behavioral problems, there was a higher incidence of parent-reported problems among children with 21-OHD than controls, including conduct problems, impulsive hyperactivity, anxiety, and hyperactivity index. CONCLUSION Parents of 21-OHD patients in Southern China were unwilling to disclose the condition of their children to the society. Masculinization and behavioral problems were prevalent among patients with 21-OHD, which highlighted the importance of psychological and social support for 21-OHD patients and their families.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pinggan Li
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Ou
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zulin Liu
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yi Sun
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingfang Song
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lele Hou
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Siqi Huang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhe Meng
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liyang Liang
- Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
18
|
Ye Y, Lu Z, Zhou W. Pheromone effects on the human hypothalamus in relation to sexual orientation and gender. HANDBOOK OF CLINICAL NEUROLOGY 2021; 182:293-306. [PMID: 34266600 DOI: 10.1016/b978-0-12-819973-2.00021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pheromones are chemicals that serve communicational purposes within a species. In most terrestrial mammals, pheromones are detected by either the olfactory epithelium or the vomeronasal organ and processed by various downstream structures including the medial amygdala and the hypothalamus to regulate motivated behaviors and endocrine responses. The search for human pheromones began in the 1970s. Whereas bioactive ligands are yet to be identified, there has been accumulating evidence that human body odors exert a range of pheromone-like effects on the recipients, including triggering innate behavioral responses, modulating endocrine levels, signaling social information, and affecting mood and cognition. In parallel, results from recent brain imaging studies suggest that body odors evoke distinct neural responses from those observed with common nonsocial odors. Two endogenous steroids androsta-4,16,- dien-3-one and estra-1,3,5(10),16-tetraen-3-ol are considered by some as candidates for human sex pheromones. The two substances produce sexually dimorphic effects on human perception, mood, and physiological arousal. Moreover, they reportedly elicit different hypothalamic response patterns in manners contingent on the recipients' sex and sexual orientation. Neuroendocrine mechanisms underlying the effects of human chemosignals are not yet clear and await future detailed analyses.
Collapse
Affiliation(s)
- Yuting Ye
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhonghua Lu
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wen Zhou
- State Key Laboratory of Brain and Cognitive Science, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
19
|
Levy DR, Sofer Y, Brumfeld V, Zilkha N, Kimchi T. The Nasopalatine Ducts Are Required for Proper Pheromone Signaling in Mice. Front Neurosci 2020; 14:585323. [PMID: 33328853 PMCID: PMC7710809 DOI: 10.3389/fnins.2020.585323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/07/2020] [Indexed: 11/25/2022] Open
Abstract
The vomeronasal organ (VNO) specializes in detection of chemosignals, mainly pheromones, which control social communication and reproduction in many mammals. These pheromones must solubilize with nasal fluids before entering the VNO, and it was suggested that they are delivered to and cleared from the VNO by active pumping. Yet, the details of this pheromone delivery process are unclear. In this study, we first constructed a high-resolution 3D morphological image of the whole adult mouse snout, by using ultra-high-resolution micro-CT. We identified a net of micro tunnels starting from the nostrils and extending around and through the VNO. These micro tunnels connect the nasal cavity with the VNO and the oral cavity via the nasopalatine ducts (NPD). Other micro tunnels connect the nasal cavity to the main olfactory epithelium. We next demonstrated that physical obstruction of the NPD severely impairs the clearance of dissolved compounds from the VNO lumen. Moreover, we found that mice with blocked NPD display alterations in chemosignaling-evoked neuronal activation in brain regions associated with the vomeronasal system. Finally, NPD-blocked male mice exhibit reduced preference for female chemosignals, and impaired social interaction behavior. Taken together, our findings indicate that the NPD in mice are connected to both the nasal and oral cavity, serving an essential role in regulating the flow of soluble chemosignals through the VNO, and are required for proper pheromone-mediated social communication.
Collapse
Affiliation(s)
- Dana Rubi Levy
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Yizhak Sofer
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Vlad Brumfeld
- Department of Structural Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Noga Zilkha
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Kimchi
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
20
|
DeAngelis RS, Hofmann HA. Neural and molecular mechanisms underlying female mate choice decisions in vertebrates. ACTA ACUST UNITED AC 2020; 223:223/17/jeb207324. [PMID: 32895328 DOI: 10.1242/jeb.207324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Female mate choice is a dynamic process that allows individuals to selectively mate with those of the opposite sex that display a preferred set of traits. Because in many species males compete with each other for fertilization opportunities, female mate choice can be a powerful agent of sexual selection, often resulting in highly conspicuous traits in males. Although the evolutionary causes and consequences of the ornamentation and behaviors displayed by males to attract mates have been well studied, embarrassingly little is known about the proximate neural mechanisms through which female choice occurs. In vertebrates, female mate choice is inherently a social behavior, and although much remains to be discovered about this process, recent evidence suggests the neural substrates and circuits underlying other fundamental social behaviors (such as pair bonding, aggression and parental care) are likely similarly recruited during mate choice. Notably, female mate choice is not static, as social and ecological environments can shape the brain and, consequently, behavior in specific ways. In this Review, we discuss how social and/or ecological influences mediate female choice and how this occurs within the brain. We then discuss our current understanding of the neural substrates underlying female mate choice, with a specific focus on those that also play a role in regulating other social behaviors. Finally, we propose several promising avenues for future research by highlighting novel model systems and new methodological approaches, which together will transform our understanding of the causes and consequences of female mate choice.
Collapse
Affiliation(s)
- Ross S DeAngelis
- Department of Integrative Biology, The University of Texas, Austin, TX 78712, USA
| | - Hans A Hofmann
- Department of Integrative Biology, The University of Texas, Austin, TX 78712, USA .,Institute for Neuroscience, The University of Texas, Austin, TX 78712, USA.,Institute for Cellular and Molecular Biology, The University of Texas, Austin, TX 78712, USA
| |
Collapse
|
21
|
Fischer EK, Nowicki JP, O'Connell LA. Evolution of affiliation: patterns of convergence from genomes to behaviour. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180242. [PMID: 31154971 DOI: 10.1098/rstb.2018.0242] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Affiliative behaviours have evolved many times across animals. Research on the mechanisms underlying affiliative behaviour demonstrates remarkable convergence across species spanning wide evolutionary distances. Shared mechanisms have been identified with genomic approaches analysing genetic variants and gene expression differences as well as neuroendocrine and molecular approaches exploring the role of hormones and signalling molecules. We review the genomic and neural basis of pair bonding and parental care across diverse taxa to shed light on mechanistic patterns that underpin the convergent evolution of affiliative behaviour. We emphasize that mechanisms underlying convergence in complex phenotypes like affiliation should be evaluated on a continuum, where signatures of convergence may vary across levels of biological organization. In particular, additional comparative studies within and across major vertebrate lineages will be essential in resolving when and why shared neural substrates are repeatedly targeted in the independent evolution of affiliation, and how similar mechanisms are evolutionarily tuned to give rise to species-specific variations in behaviour. This article is part of the theme issue 'Convergent evolution in the genomics era: new insights and directions'.
Collapse
Affiliation(s)
- Eva K Fischer
- Department of Biology, Stanford University , Stanford, CA 95305 , USA
| | - Jessica P Nowicki
- Department of Biology, Stanford University , Stanford, CA 95305 , USA
| | | |
Collapse
|
22
|
Zhang YH, Tang MM, Guo X, Gao XR, Zhang JH, Zhang JX. Associative learning is necessary for airborne pheromones to activate sexual arousal-linked brain areas of female rats. Behav Ecol Sociobiol 2019. [DOI: 10.1007/s00265-019-2685-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
23
|
Sexually Dimorphic Control of Parenting Behavior by the Medial Amygdala. Cell 2019; 176:1206-1221.e18. [PMID: 30773317 DOI: 10.1016/j.cell.2019.01.024] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/29/2018] [Accepted: 01/09/2019] [Indexed: 11/21/2022]
Abstract
Social behaviors, including behaviors directed toward young offspring, exhibit striking sex differences. Understanding how these sexually dimorphic behaviors are regulated at the level of circuits and transcriptomes will provide insights into neural mechanisms of sex-specific behaviors. Here, we uncover a sexually dimorphic role of the medial amygdala (MeA) in governing parental and infanticidal behaviors. Contrary to traditional views, activation of GABAergic neurons in the MeA promotes parental behavior in females, while activation of this population in males differentially promotes parental versus infanticidal behavior in an activity-level-dependent manner. Through single-cell transcriptomic analysis, we found that molecular sex differences in the MeA are specifically represented in GABAergic neurons. Collectively, these results establish crucial roles for the MeA as a key node in the neural circuitry underlying pup-directed behaviors and provide important insight into the connection between sex differences across transcriptomes, cells, and circuits in regulating sexually dimorphic behavior.
Collapse
|
24
|
Hiura LC, Ophir AG. Interactions of sex and early life social experiences at two developmental stages shape nonapeptide receptor profiles. Integr Zool 2019; 13:745-760. [PMID: 29851289 DOI: 10.1111/1749-4877.12338] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Early life social experiences are critical to behavioral and cognitive development, and can have a tremendous influence on developing social phenotypes. Most work has focused on outcomes of experiences at a single stage of development (e.g. perinatal or post-weaning). Few studies have assessed the impact of social experience at multiple developmental stages and across sex. Oxytocin and vasopressin are profoundly important for modulating social behavior and these nonapeptide systems are highly sensitive to developmental social experience, particularly in brain areas important for social behavior. We investigated whether oxytocin receptor (OTR) and vasopressin receptor (V1aR) distributions of prairie voles (Microtus ochrogaster) change as a function of parental composition within the natal nest or social composition after weaning. We raised pups either in the presence or absence of their fathers. At weaning, offspring were housed either individually or with a same-sex sibling. We also examined whether changes in receptor distributions are sexually dimorphic because the impact of the developmental environment on the nonapeptide system could be sex-dependent. We found that differences in nonapeptide receptor expression were region-specific, sex-specific and rearing condition-specific, indicating a high level of complexity in the ways that early life experiences shape the social brain. We found many more differences in V1aR density compared to OTR density, indicating that nonapeptide receptors demonstrate differential levels of neural plasticity and sensitivity to environmental and biological variables. Our data highlight that critical factors including biological sex and multiple experiences across the developmental continuum interact in complex ways to shape the social brain.
Collapse
Affiliation(s)
- Lisa C Hiura
- Department of Psychology, Cornell University, Ithaca, New York, USA
| | | |
Collapse
|
25
|
Neural coding of sex-specific social information in the mouse brain. Curr Opin Neurobiol 2018; 53:120-130. [DOI: 10.1016/j.conb.2018.07.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/11/2018] [Accepted: 07/11/2018] [Indexed: 12/19/2022]
|
26
|
Daghfous G, Auclair F, Clotten F, Létourneau JL, Atallah E, Millette JP, Derjean D, Robitaille R, Zielinski BS, Dubuc R. GABAergic modulation of olfactomotor transmission in lampreys. PLoS Biol 2018; 16:e2005512. [PMID: 30286079 PMCID: PMC6191151 DOI: 10.1371/journal.pbio.2005512] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/16/2018] [Accepted: 09/18/2018] [Indexed: 12/21/2022] Open
Abstract
Odor-guided behaviors, including homing, predator avoidance, or food and mate searching, are ubiquitous in animals. It is only recently that the neural substrate underlying olfactomotor behaviors in vertebrates was uncovered in lampreys. It consists of a neural pathway extending from the medial part of the olfactory bulb (medOB) to locomotor control centers in the brainstem via a single relay in the caudal diencephalon. This hardwired olfactomotor pathway is present throughout life and may be responsible for the olfactory-induced motor behaviors seen at all life stages. We investigated modulatory mechanisms acting on this pathway by conducting anatomical (tract tracing and immunohistochemistry) and physiological (intracellular recordings and calcium imaging) experiments on lamprey brain preparations. We show that the GABAergic circuitry of the olfactory bulb (OB) acts as a gatekeeper of this hardwired sensorimotor pathway. We also demonstrate the presence of a novel olfactomotor pathway that originates in the non-medOB and consists of a projection to the lateral pallium (LPal) that, in turn, projects to the caudal diencephalon and to the mesencephalic locomotor region (MLR). Our results indicate that olfactory inputs can induce behavioral responses by activating brain locomotor centers via two distinct pathways that are strongly modulated by GABA in the OB. The existence of segregated olfactory subsystems in lampreys suggests that the organization of the olfactory system in functional clusters may be a common ancestral trait of vertebrates.
Collapse
Affiliation(s)
- Gheylen Daghfous
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
- Groupe de Recherche en Activité Physique Adaptée, Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada
| | - François Auclair
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
| | - Felix Clotten
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Luc Létourneau
- Groupe de Recherche en Activité Physique Adaptée, Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Elias Atallah
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Patrick Millette
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
| | - Dominique Derjean
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
- Groupe de Recherche en Activité Physique Adaptée, Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada
| | - Richard Robitaille
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
| | - Barbara S. Zielinski
- Department of Biological Sciences, University of Windsor, Windsor, Ontario, Canada
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, Ontario, Canada
| | - Réjean Dubuc
- Groupe de Recherche sur le Système Nerveux Central, Département de neurosciences, Université de Montréal, Montréal, Québec, Canada
- Groupe de Recherche en Activité Physique Adaptée, Département des sciences de l'activité physique, Université du Québec à Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
27
|
Hilbert ZA, Kim DH. PDF-1 neuropeptide signaling regulates sexually dimorphic gene expression in shared sensory neurons of C. elegans. eLife 2018; 7:36547. [PMID: 30024377 PMCID: PMC6053303 DOI: 10.7554/elife.36547] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022] Open
Abstract
Sexually dimorphic behaviors are a feature common to species across the animal kingdom, however how such behaviors are generated from mostly sex-shared nervous systems is not well understood. Building on our previous work which described the sexually dimorphic expression of a neuroendocrine ligand, DAF-7, and its role in behavioral decision-making in C. elegans (Hilbert and Kim, 2017), we show here that sex-specific expression of daf-7 is regulated by another neuroendocrine ligand, Pigment Dispersing Factor (PDF-1), which has previously been implicated in regulating male-specific behavior (Barrios et al., 2012). Our analysis revealed that PDF-1 signaling acts sex- and cell-specifically in the ASJ neurons to regulate the expression of daf-7, and we show that differences in PDFR-1 receptor activity account for the sex-specific effects of this pathway. Our data suggest that modulation of the sex-shared nervous system by a cascade of neuroendocrine signals can shape sexually dimorphic behaviors.
Collapse
Affiliation(s)
- Zoë A Hilbert
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| | - Dennis H Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
28
|
Kohl J, Dulac C. Neural control of parental behaviors. Curr Opin Neurobiol 2018; 49:116-122. [PMID: 29482085 DOI: 10.1016/j.conb.2018.02.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/27/2017] [Accepted: 02/06/2018] [Indexed: 11/18/2022]
Abstract
Parenting is a multicomponent social behavior that is essential for the survival of offspring in many species. Despite extensive characterization of individual brain areas involved in parental care, we do not fully understand how discrete aspects of this behavior are orchestrated at the neural circuit level. Recent progress in identifying genetically specified neuronal populations critical for parenting, and the use of genetic and viral tools for circuit-cracking now allow us to deconstruct the underlying circuitry and, thus, to elucidate how different aspects of parental care are controlled. Here we review the latest advances, outline possible organizational principles of parental circuits and discuss future challenges.
Collapse
Affiliation(s)
- Johannes Kohl
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
29
|
Albin RL. Tourette syndrome: a disorder of the social decision-making network. Brain 2018; 141:332-347. [PMID: 29053770 PMCID: PMC5837580 DOI: 10.1093/brain/awx204] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/08/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022] Open
Abstract
Tourette syndrome is a common neurodevelopmental disorder defined by characteristic involuntary movements, tics, with both motor and phonic components. Tourette syndrome is usually conceptualized as a basal ganglia disorder, with an emphasis on striatal dysfunction. While considerable evidence is consistent with these concepts, imaging data suggest diffuse functional and structural abnormalities in Tourette syndrome brain. Tourette syndrome exhibits features that are difficult to explain solely based on basal ganglia circuit dysfunctions. These features include the natural history of tic expression, with typical onset of tics around ages 5 to 7 years and exacerbation during the peri-pubertal years, marked sex disparity with higher male prevalence, and the characteristic distribution of tics. The latter are usually repetitive, somewhat stereotyped involuntary eye, facial and head movements, and phonations. A major functional role of eye, face, and head movements is social signalling. Prior work in social neuroscience identified a phylogenetically conserved network of sexually dimorphic subcortical nuclei, the Social Behaviour Network, mediating many social behaviours. Social behaviour network function is modulated developmentally by gonadal steroids and social behaviour network outputs are stereotyped sex and species specific behaviours. In 2011 O'Connell and Hofmann proposed that the social behaviour network interdigitates with the basal ganglia to form a greater network, the social decision-making network. The social decision-making network may have two functionally complementary limbs: the basal ganglia component responsible for evaluation of socially relevant stimuli and actions with the social behaviour network component responsible for the performance of social acts. Social decision-making network dysfunction can explain major features of the neurobiology of Tourette syndrome. Tourette syndrome may be a disorder of social communication resulting from developmental abnormalities at several levels of the social decision-making network. The social decision-making network dysfunction hypothesis suggests new avenues for research in Tourette syndrome and new potential therapeutic targets.
Collapse
Affiliation(s)
- Roger L Albin
- Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA
- Neurology Service and GRECC, VAAAHS, Ann Arbor, MI, 48105, USA
- University of Michigan Morris K. Udall Parkinson’s Disease Research Center, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
30
|
Chung AS, Miller SM, Sun Y, Xu X, Zweifel LS. Sexual congruency in the connectome and translatome of VTA dopamine neurons. Sci Rep 2017; 7:11120. [PMID: 28894175 PMCID: PMC5593921 DOI: 10.1038/s41598-017-11478-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/22/2017] [Indexed: 01/13/2023] Open
Abstract
The ventral tegmental area (VTA) dopamine system is important for reward, motivation, emotion, learning, and memory. Dysfunctions in the dopamine system are linked to multiple neurological and neuropsychiatric disorders, many of which present with sex differences. Little is known about the extent of heterogeneity in the basic organization of VTA dopamine neurons with regard to sex. Here, we characterized the cell-specific connectivity of VTA dopamine neurons, their mRNA translational profile, and basic electrophysiological characteristics in a common strain of mice. We found no major differences in these metrics, except for differential expression of a Y-chromosome associated mRNA transcript, Eif2s3y, and the X-linked, X-inactivation transcript Xist. Of note, Xist transcript was significantly enriched in dopamine neurons, suggesting tight regulation of X-linked gene expression to ensure sexual congruency. These data indicate that the features that make dopamine neurons unique are highly concordant and not a principal source of sexual dimorphism.
Collapse
Affiliation(s)
- Amanda S Chung
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - Samara M Miller
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA
| | - Yanjun Sun
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Larry S Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA, 98195, USA. .,Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
31
|
Experience-Dependent Plasticity Drives Individual Differences in Pheromone-Sensing Neurons. Neuron 2017; 91:878-892. [PMID: 27537487 DOI: 10.1016/j.neuron.2016.07.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 03/30/2016] [Accepted: 07/06/2016] [Indexed: 02/03/2023]
Abstract
Different individuals exhibit distinct behaviors, but studying the neuronal basis of individuality is a daunting challenge. Here, we considered this question in the vomeronasal organ, a pheromone-detecting epithelium containing hundreds of distinct neuronal types. Using light-sheet microscopy, we characterized in each animal the abundance of 17 physiologically defined types, altogether recording from half a million sensory neurons. Inter-animal differences were much larger than predicted by chance, and different physiological cell types showed distinct patterns of variability. One neuronal type was present in males and nearly absent in females. Surprisingly, this apparent sexual dimorphism was generated by plasticity, as exposure to female scents or single ligands led to both the elimination of this cell type and alterations in olfactory behavior. That an all-or-none apparent sex difference in neuronal types is controlled by experience-even in a sensory system devoted to "innate" behaviors-highlights the extraordinary role of "nurture" in neural individuality.
Collapse
|
32
|
Hurtado-López J, Ramirez-Moreno DF, Sejnowski TJ. Decision-making neural circuits mediating social behaviors : An attractor network model. J Comput Neurosci 2017; 43:127-142. [PMID: 28660531 DOI: 10.1007/s10827-017-0654-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 11/24/2022]
Abstract
We propose a mathematical model of a continuous attractor network that controls social behaviors. The model is examined with bifurcation analysis and computer simulations. The results show that the model exhibits stable steady states and thresholds for steady state transitions corresponding to some experimentally observed behaviors, such as aggression control. The performance of the model and the relation with experimental evidence are discussed.
Collapse
Affiliation(s)
- Julián Hurtado-López
- Department of Mathematics, Universidad Autónoma de Occidente, Cll 25 No. 115-85 Km 2 vía Cali-Jamundí, 760030, Cali, Colombia.
| | | | - Terrence J Sejnowski
- Howard Hughes Medical Institute, the Salk Institute for Biological Studies, La Jolla, California, 92037, USA
| |
Collapse
|
33
|
Razzaghy-Azar M, Karimi S, Shirazi E. Gender Identity in Patients with Congenital Adrenal Hyperplasia. Int J Endocrinol Metab 2017; 15:e12537. [PMID: 29201068 PMCID: PMC5701969 DOI: 10.5812/ijem.12537] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 05/23/2017] [Accepted: 07/02/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Sex assignment in infancy for patients with disorder of sex development (DSD) is a challenging problem. Some of the patients with congenital adrenal hyperplasia (CAH) have DSD that may affect their gender identity. OBJECTIVES The study aimed to assess gender identity in patients with CAH. METHODS In this study, 52 patients with CAH, including 22 prepubertal children and 30 adolescents and adults, were assessed using two separate gender identity questionnaires for children and adults based on the criteria of diagnostic and statistical manual of mental disorders, 5th edition. RESULTS In the children group, compatibility was seen between gender identity and rearing gender. In the adult group, there were three cases of mismatching between gender identity and sex assignment composed of two females with poor control and one male with good control with 21-hydroxylase deficiency (21-OHD). Three girls with 11-hydroxylase deficiency (11-OHD) were reared as boy. Two of them with late diagnosis at 5 and 6 years of age had pseudoprecocious puberty. Parents and children did not accept to change the gender. One of them is 36 years old now, is depressed and unsatisfied with her gender, another girl is still child and has male sexual identity. One girl with 11-OHD and early diagnosis at birth with Prader 5 virilization but with good hormonal control was changed to female gender at 12 years of age when female sexual characteristics appeared; she is 34-years-old now, married, and with two children, and she is satisfied with her gender. CONCLUSIONS In patients with CAH, gender identity disorder is a rare finding. Hormonal control, social, familial, and religious beliefs have impacts on gender identity of these patients.
Collapse
Affiliation(s)
- Maryam Razzaghy-Azar
- H. Ali-Asghar Children’s Hospital, Iran University of Medical Sciences, Tehran, Iran
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sakineh Karimi
- H. Ali-Asghar Children’s Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Shirazi
- H. Ali-Asghar Children’s Hospital, Iran University of Medical Sciences, Tehran, Iran
- Mental Health Research Center, Tehran Institute of Psychiatry-School of Behavioral Sciences and Mental Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Krzyszczyk E, Patterson EM, Stanton MA, Mann J. The transition to independence: sex differences in social and behavioural development of wild bottlenose dolphins. Anim Behav 2017. [DOI: 10.1016/j.anbehav.2017.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
35
|
Walker DM, Gore AC. Epigenetic impacts of endocrine disruptors in the brain. Front Neuroendocrinol 2017; 44:1-26. [PMID: 27663243 PMCID: PMC5429819 DOI: 10.1016/j.yfrne.2016.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/05/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
The acquisition of reproductive competence is organized and activated by steroid hormones acting upon the hypothalamus during critical windows of development. This review describes the potential role of epigenetic processes, particularly DNA methylation, in the regulation of sexual differentiation of the hypothalamus by hormones. We examine disruption of these processes by endocrine-disrupting chemicals (EDCs) in an age-, sex-, and region-specific manner, focusing on how perinatal EDCs act through epigenetic mechanisms to reprogram DNA methylation and sex steroid hormone receptor expression throughout life. These receptors are necessary for brain sexual differentiation and their altered expression may underlie disrupted reproductive physiology and behavior. Finally, we review the literature on histone modifications and non-coding RNA involvement in brain sexual differentiation and their perturbation by EDCs. By putting these data into a sex and developmental context we conclude that perinatal EDC exposure alters the developmental trajectory of reproductive neuroendocrine systems in a sex-specific manner.
Collapse
Affiliation(s)
- Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1065, New York, NY 10029, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, and The University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
36
|
Capturing and Manipulating Activated Neuronal Ensembles with CANE Delineates a Hypothalamic Social-Fear Circuit. Neuron 2016; 92:739-753. [PMID: 27974160 DOI: 10.1016/j.neuron.2016.10.015] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/22/2016] [Accepted: 10/03/2016] [Indexed: 01/03/2023]
Abstract
We developed a technology (capturing activated neuronal ensembles [CANE]) to label, manipulate, and transsynaptically trace neural circuits that are transiently activated in behavioral contexts with high efficiency and temporal precision. CANE consists of a knockin mouse and engineered viruses designed to specifically infect activated neurons. Using CANE, we selectively labeled neurons that were activated by either fearful or aggressive social encounters in a hypothalamic subnucleus previously known as a locus for aggression, and discovered that social-fear and aggression neurons are intermixed but largely distinct. Optogenetic stimulation of CANE-captured social-fear neurons (SFNs) is sufficient to evoke fear-like behaviors in normal social contexts, whereas silencing SFNs resulted in reduced social avoidance. CANE-based mapping of axonal projections and presynaptic inputs to SFNs further revealed a highly distributed and recurrent neural network. CANE is a broadly applicable technology for dissecting causality and connectivity of spatially intermingled but functionally distinct ensembles.
Collapse
|
37
|
Rezával C, Pattnaik S, Pavlou HJ, Nojima T, Brüggemeier B, D'Souza LAD, Dweck HKM, Goodwin SF. Activation of Latent Courtship Circuitry in the Brain of Drosophila Females Induces Male-like Behaviors. Curr Biol 2016; 26:2508-2515. [PMID: 27568592 PMCID: PMC5049544 DOI: 10.1016/j.cub.2016.07.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/13/2016] [Accepted: 07/12/2016] [Indexed: 12/02/2022]
Abstract
Courtship in Drosophila melanogaster offers a powerful experimental paradigm for the study of innate sexually dimorphic behaviors [1, 2]. Fruit fly males exhibit an elaborate courtship display toward a potential mate [1, 2]. Females never actively court males, but their response to the male’s display determines whether mating will actually occur. Sex-specific behaviors are hardwired into the nervous system via the actions of the sex determination genes doublesex (dsx) and fruitless (fru) [1]. Activation of male-specific dsx/fru+ P1 neurons in the brain initiates the male’s courtship display [3, 4], suggesting that neurons unique to males trigger this sex-specific behavior. In females, dsx+ neurons play a pivotal role in sexual receptivity and post-mating behaviors [1, 2, 5, 6, 7, 8, 9]. Yet it is still unclear how dsx+ neurons and dimorphisms in these circuits give rise to the different behaviors displayed by males and females. Here, we manipulated the function of dsx+ neurons in the female brain to investigate higher-order neurons that drive female behaviors. Surprisingly, we found that activation of female dsx+ neurons in the brain induces females to behave like males by promoting male-typical courtship behaviors. Activated females display courtship toward conspecific males or females, as well other Drosophila species. We uncovered specific dsx+ neurons critical for driving male courtship and identified pheromones that trigger such behaviors in activated females. While male courtship behavior was thought to arise from male-specific central neurons, our study shows that the female brain is equipped with latent courtship circuitry capable of inducing this male-specific behavioral program. Activation of brain dsx-pC1 neurons promote male-like courtship in females Activated females court conspecific males and females and other Drosophila species Methyl pheromones trigger male courtship behaviors in activated females The female brain is equipped with latent circuitry underlying male-like behavior
Collapse
Affiliation(s)
- Carolina Rezával
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK.
| | - Siddharth Pattnaik
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Hania J Pavlou
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Tetsuya Nojima
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Birgit Brüggemeier
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Luis A D D'Souza
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Hany K M Dweck
- Department of Evolutionary Neuroethology, Max Planck Institute for Chemical Ecology, 07745 Jena, Germany
| | - Stephen F Goodwin
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| |
Collapse
|
38
|
Zhou C, Franconville R, Vaughan AG, Robinett CC, Jayaraman V, Baker BS. Central neural circuitry mediating courtship song perception in male Drosophila. eLife 2015; 4. [PMID: 26390382 PMCID: PMC4575990 DOI: 10.7554/elife.08477] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 08/24/2015] [Indexed: 11/13/2022] Open
Abstract
Animals use acoustic signals across a variety of social behaviors, particularly courtship. In Drosophila, song is detected by antennal mechanosensory neurons and further processed by second-order aPN1/aLN(al) neurons. However, little is known about the central pathways mediating courtship hearing. In this study, we identified a male-specific pathway for courtship hearing via third-order ventrolateral protocerebrum Projection Neuron 1 (vPN1) neurons and fourth-order pC1 neurons. Genetic inactivation of vPN1 or pC1 disrupts song-induced male-chaining behavior. Calcium imaging reveals that vPN1 responds preferentially to pulse song with long inter-pulse intervals (IPIs), while pC1 responses to pulse song closely match the behavioral chaining responses at different IPIs. Moreover, genetic activation of either vPN1 or pC1 induced courtship chaining, mimicking the behavioral response to song. These results outline the aPN1-vPN1-pC1 pathway as a labeled line for the processing and transformation of courtship song in males. DOI:http://dx.doi.org/10.7554/eLife.08477.001 The seemingly simple fruit fly engages in an intricate courtship ritual before it mates. Male flies use their wings to ‘sing’ a complex song that makes females more willing to mate. The song also encourages nearby males to start courting, and these males may then intervene to compete for the female. Each species of fruit fly has its own song, and it is important for both males and females to detect the right song. The sounds of the courtship song are detected by vibration-sensitive neurons on the flies' antennae. These neurons send signals to the fly's brain. But little is known about how this information is then processed, or how information about the song can be integrated with other courtship cues. Zhou et al. have now identified a pathway of neurons in male flies that is responsible for hearing the courtship song. This pathway stretches from the antennae to neurons deep within the brain. These neural pathways are different in males and females, suggesting that the two sexes use different circuits of neurons for hearing courtship songs. Zhou et al. then used genetic techniques to show that males need every neuron in this pathway to hear courtship songs. Further experiments revealed that stimulating the ‘deep layer’ neurons caused male flies to respond as if they are hearing the courtship song. These neurons are likely to integrate the song with information from other senses and may encode a general signal for arousal. These findings now pave the way to deepen our understanding of how information from different senses—for example, courtship songs, visual cues, and pheromones—can be integrated to drive specific behaviors. The next challenge is to explore how species-specific songs are detected and recognized, a goal that has yet to be achieved in any species. DOI:http://dx.doi.org/10.7554/eLife.08477.002
Collapse
Affiliation(s)
- Chuan Zhou
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Romain Franconville
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | | | - Carmen C Robinett
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Vivek Jayaraman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| | - Bruce S Baker
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, United States
| |
Collapse
|
39
|
Jahan MR, Kokubu K, Islam MN, Matsuo C, Yanai A, Wroblewski G, Fujinaga R, Shinoda K. Species differences in androgen receptor expression in the medial preoptic and anterior hypothalamic areas of adult male and female rodents. Neuroscience 2014; 284:943-961. [PMID: 25446364 DOI: 10.1016/j.neuroscience.2014.11.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 11/03/2014] [Accepted: 11/04/2014] [Indexed: 11/25/2022]
Abstract
The medial preoptic and anterior hypothalamic areas (MPO/AH) are important androgen targets regulating homeostasis, neuroendocrinology and circadian rhythm as well as instinctive and sociosexual behaviors. Although species differences between rats and mice have been pointed out in terms of morphology and physiology, detailed distributions of androgen receptor (AR) have never been compared between the two rodents. In the present study, AR distribution was examined immunohistochemically in serial sections of the MPO/AH and compared for adult rats and mice. Western blotting and immunohistochemistry clearly demonstrated that AR expression in the brain was stronger in mice than in rats and was stronger in males than in females. In addition, we found (1) an "obliquely elongated calbindin-ir cell island" in mice medial preoptic nucleus (MPN) expressed AR intensely, as well as the sexually dimorphic nucleus in the MPN (SDN-MPN) in rats, strongly supporting a "putative SDN-MPN" previously proposed in mice; (2) AR expression in the suprachiasmatic nucleus (SCN) was much more prominent in mice than in rats and differed in localization between the two species; (3) a mouse-specific AR-ir cell cluster was newly identified as the "tear drop nucleus (TDN)", with male-dominant sexual dimorphism; and (4) two rat-specific AR-ir cell clusters were also newly identified as the "rostral and caudal nebular islands", with male-dominant sexual dimorphism. The present results may provide basic morphological evidence underlying species differences in androgen-modified psychological, physiological and endocrinergic responses. Above all, the findings of the mouse-specific TDN and differing AR expression in the SCN might explain not only species difference in gonadal modification of circadian rhythm, but also distinct structural bases in the context of transduction of SCN oscillation. The current study could also serve as a caution that data on androgen-sensitive functions obtained from one species should not always be directly applied to others among rodents.
Collapse
Affiliation(s)
- M R Jahan
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - K Kokubu
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - Md N Islam
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - C Matsuo
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - A Yanai
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - G Wroblewski
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - R Fujinaga
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan
| | - K Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| |
Collapse
|
40
|
|
41
|
Chalfin L, Dayan M, Levy DR, Austad SN, Miller RA, Iraqi FA, Dulac C, Kimchi T. Mapping ecologically relevant social behaviours by gene knockout in wild mice. Nat Commun 2014; 5:4569. [DOI: 10.1038/ncomms5569] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 07/01/2014] [Indexed: 11/10/2022] Open
|
42
|
The interplay between reproductive social stimuli and adult olfactory bulb neurogenesis. Neural Plast 2014; 2014:497657. [PMID: 25140258 PMCID: PMC4130132 DOI: 10.1155/2014/497657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 06/19/2014] [Indexed: 12/11/2022] Open
Abstract
Adult neurogenesis is a striking form of structural plasticity that adapts the brain to the changing world. Accordingly, new neuron production is involved in cognitive functions, such as memory, learning, and pattern separation. Recent data in rodents indicate a close link between adult neurogenesis and reproductive social behavior. This provides a key to unravel the functional meaning of adult neurogenesis in biological relevant contexts and, in parallel, opens new perspectives to explore the way the brain is processing social stimuli. In this paper we will summarize some of the major achievements on cues and mechanisms modulating adult neurogenesis during social behaviors related to reproduction and possible role/s played by olfactory newborn neurons in this context. We will point out that newborn interneurons in the accessory olfactory bulb (AOB) represent a privileged cellular target for social stimuli that elicit reproductive behaviors and that such cues modulate adult neurogenesis at two different levels increasing both proliferation of neuronal progenitors in the germinative regions and integration of newborn neurons into functional circuits. This dual mechanism provides fresh neurons that can be involved in critical activities for the individual fitness, that is, the processing of social stimuli driving the parental behavior and partner recognition.
Collapse
|
43
|
Abstract
Animal–animal recognition within, and across species, is essential for predator avoidance and social interactions. Despite its essential role in orchestrating responses to animal cues, basic principles of information processing by the vomeronasal system are still unknown. The medial amygdala (MeA) occupies a central position in the vomeronasal pathway, upstream of hypothalamic centers dedicated to defensive and social responses. We have characterized sensory responses in the mouse MeA and uncovered emergent properties that shed new light onto the transformation of vomeronasal information into sex- and species-specific responses. In particular, we show that the MeA displays a degree of stimulus selectivity and a striking sexually dimorphic sensory representation that are not observed in the upstream relay of the accessory olfactory bulb (AOB). Furthermore, our results demonstrate that the development of sexually dimorphic circuits in the MeA requires steroid signaling near the time of puberty to organize the functional representation of sensory stimuli. DOI:http://dx.doi.org/10.7554/eLife.02743.001 Many animals emit and detect chemicals known as pheromones to communicate with other members of their own species. Animals also rely on chemical signals from other species to warn them, for example, that a predator is nearby. Many of these chemical signals—which are present in sweat, tears, urine, and saliva—are detected by a structure called the vomeronasal organ, which is located at the base of the nasal cavity. When this organ detects a particular chemical signal, it broadcasts this information to a network of brain regions that generates an appropriate behavioral response. Two structures within this network, the accessory olfactory bulb and the medial amygdala, play an important role in modifying this signal before it reaches its final destination—a region of the brain called the hypothalamus. Activation of the hypothalamus by the signal triggers changes in the animal's behavior. Although the anatomical details of this pathway have been widely studied, it is not clear how information is actually transmitted along it. Now, Bergan et al. have provided insights into this process by recording signals in the brains of anesthetized mice exposed to specific stimuli. Whereas neurons in the accessory olfactory bulb responded similarly in male and female mice, those in the medial amygdala showed a preference for female urine in male mice, and a preference for male urine in the case of females. This is the first direct demonstration of differences in sensory processing in the brains of male and female mammals. These differences are thought to result from the actions of sex hormones, particularly estrogen, on brain circuits during development. Consistent with this, neurons in the medial amygdala of male mice with reduced levels of estrogen showed a reduced preference for female urine compared to control males. Similarly, female mice that had been previously exposed to high levels of estrogen as pups showed a reduced preference for male urine compared to controls. In addition to increasing understanding of how chemical signals—including pheromones—influence the responses of rodents to other animals, the work of Bergan et al. has provided clues to the neural mechanisms that underlie sex-specific differences in behaviors. DOI:http://dx.doi.org/10.7554/eLife.02743.002
Collapse
Affiliation(s)
- Joseph F Bergan
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| | - Yoram Ben-Shaul
- School of Medicine, Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Catherine Dulac
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| |
Collapse
|
44
|
Bergan JF, Ben-Shaul Y, Dulac C. Sex-specific processing of social cues in the medial amygdala. eLife 2014. [PMID: 24894465 DOI: 10.7554/elife.02743.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Animal-animal recognition within, and across species, is essential for predator avoidance and social interactions. Despite its essential role in orchestrating responses to animal cues, basic principles of information processing by the vomeronasal system are still unknown. The medial amygdala (MeA) occupies a central position in the vomeronasal pathway, upstream of hypothalamic centers dedicated to defensive and social responses. We have characterized sensory responses in the mouse MeA and uncovered emergent properties that shed new light onto the transformation of vomeronasal information into sex- and species-specific responses. In particular, we show that the MeA displays a degree of stimulus selectivity and a striking sexually dimorphic sensory representation that are not observed in the upstream relay of the accessory olfactory bulb (AOB). Furthermore, our results demonstrate that the development of sexually dimorphic circuits in the MeA requires steroid signaling near the time of puberty to organize the functional representation of sensory stimuli.DOI: http://dx.doi.org/10.7554/eLife.02743.001.
Collapse
Affiliation(s)
- Joseph F Bergan
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| | - Yoram Ben-Shaul
- School of Medicine, Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Catherine Dulac
- Molecular and Cellular Biology, Howard Hughes Medical Institute, Harvard University, Cambridge, United States
| |
Collapse
|
45
|
Canonical transient receptor potential channel 2 (TRPC2): old name-new games. Importance in regulating of rat thyroid cell physiology. Pflugers Arch 2014; 466:2025-34. [PMID: 24722829 DOI: 10.1007/s00424-014-1509-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 03/25/2014] [Accepted: 03/26/2014] [Indexed: 12/14/2022]
Abstract
In addition to the TSH-cyclic AMP signalling pathway, calcium signalling is of crucial importance in thyroid cells. Although the importance of calcium signalling has been thoroughly investigated for several decades, the nature of the calcium channels involved in signalling is unknown. In a recent series of investigations using the well-studied rat thyroid FRTL-5 cell line, we showed that these cells exclusively express the transient receptor potential canonical 2 (TRPC2) channel. Our results suggested that the TRPC2 channel is of significant importance in regulating thyroid cell function. These investigations were the first to show that thyroid cells express a member of the TRPC family of ion channels. In this review, we will describe the importance of the TRPC2 channel in regulating TSH receptor expression, thyroglobulin maturation, intracellular calcium and iodide homeostasis and that the channel also regulates thyroid cell proliferation.
Collapse
|
46
|
Kim GW, Jeong GW. A comparative study of brain activation patterns associated with sexual arousal between males and females using 3.0-T functional magnetic resonance imaging. Sex Health 2014; 11:11-6. [DOI: 10.1071/sh13127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 10/01/2013] [Indexed: 02/01/2023]
Abstract
Background In contrast to the previous studies using a 1.5-T magnetic resonance imaging system, our study was performed on a higher magnetic field strength, 3.0 T, to gain more valuable information on the functional brain anatomy associated with visual sexual arousal for discriminating the gender difference by increasing the detection power of brain activation. Methods: Twenty-four healthy subjects consisting of 12 males and 12 females underwent functional magnetic resonance imaging examination for this study. Brain activity was measured while viewing erotic videos. Results: The predominant activation areas observed in males as compared with females included the hypothalamus, the globus pallidus, the head of the caudate nucleus, the parahippocampal gyrus, the amygdala and the septal area, whereas the predominant activation in females was observed in the anterior cingulate gyrus and the putamen. Conclusion: Our findings suggest that the brain activation patterns associated with visual sexual arousal are specific to gender. This gender difference in brain activation patterns is more remarkable at higher magnet field (3.0 T) than at 1.5 T.
Collapse
|
47
|
Kohl J, Ostrovsky AD, Frechter S, Jefferis GSXE. A bidirectional circuit switch reroutes pheromone signals in male and female brains. Cell 2013; 155:1610-23. [PMID: 24360281 PMCID: PMC3898676 DOI: 10.1016/j.cell.2013.11.025] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/02/2013] [Accepted: 11/08/2013] [Indexed: 02/06/2023]
Abstract
The Drosophila sex pheromone cVA elicits different behaviors in males and females. First- and second-order olfactory neurons show identical pheromone responses, suggesting that sex genes differentially wire circuits deeper in the brain. Using in vivo whole-cell electrophysiology, we now show that two clusters of third-order olfactory neurons have dimorphic pheromone responses. One cluster responds in females; the other responds in males. These clusters are present in both sexes and share a common input pathway, but sex-specific wiring reroutes pheromone information. Regulating dendritic position, the fruitless transcription factor both connects the male-responsive cluster and disconnects the female-responsive cluster from pheromone input. Selective masculinization of third-order neurons transforms their morphology and pheromone responses, demonstrating that circuits can be functionally rewired by the cell-autonomous action of a switch gene. This bidirectional switch, analogous to an electrical changeover switch, provides a simple circuit logic to activate different behaviors in males and females.
Collapse
Affiliation(s)
- Johannes Kohl
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Aaron D Ostrovsky
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Shahar Frechter
- Division of Neurobiology, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | |
Collapse
|
48
|
Regulation of onset of female mating and sex pheromone production by juvenile hormone in Drosophila melanogaster. Proc Natl Acad Sci U S A 2013; 110:18321-6. [PMID: 24145432 DOI: 10.1073/pnas.1318119110] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Juvenile hormone (JH) coordinates timing of female reproductive maturation in most insects. In Drosophila melanogaster, JH plays roles in both mating and egg maturation. However, very little is known about the molecular pathways associated with mating. Our behavioral analysis of females genetically lacking the corpora allata, the glands that produce JH, showed that they were courted less by males and mated later than control females. Application of the JH mimic, methoprene, to the allatectomized females just after eclosion rescued both the male courtship and the mating delay. Our studies of the null mutants of the JH receptors, Methoprene tolerant (Met) and germ cell-expressed (gce), showed that lack of Met in Met(27) females delayed the onset of mating, whereas lack of Gce had little effect. The Met(27) females were shown to be more attractive but less behaviorally receptive to copulation attempts. The behavioral but not the attractiveness phenotype was rescued by the Met genomic transgene. Analysis of the female cuticular hydrocarbon profiles showed that corpora allata ablation caused a delay in production of the major female-specific sex pheromones (the 7,11-C27 and -C29 dienes) and a change in the cuticular hydrocarbon blend. In the Met(27) null mutant, by 48 h, the major C27 diene was greatly increased relative to wild type. In contrast, the gce(2.5k) null mutant females were courted similarly to control females despite changes in certain cuticular hydrocarbons. Our findings indicate that JH acts primarily via Met to modulate the timing of onset of female sex pheromone production and mating.
Collapse
|
49
|
Balthazart J, de Meaultsart CC, Ball GF, Cornil CA. Distinct neuroendocrine mechanisms control neural activity underlying sex differences in sexual motivation and performance. Eur J Neurosci 2013; 37:735-42. [PMID: 23282041 PMCID: PMC3594409 DOI: 10.1111/ejn.12102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 11/12/2012] [Accepted: 11/19/2012] [Indexed: 11/28/2022]
Abstract
Sexual behavior can be usefully parsed into an appetitive and a consummatory component. Both appetitive and consummatory male-typical sexual behaviors (respectively, ASB and CSB) are activated in male Japanese quail by testosterone (T) acting in the medial preoptic nucleus (POM), but never observed in females. This sex difference is based on a demasculinization (= organizational effect) by estradiol during embryonic life for CSB, but a differential activation by T in adulthood for ASB. Males expressing rhythmic cloacal sphincter movements (RCSMs; a form of ASB) or allowed to copulate display increased Fos expression in POM. We investigated Fos brain responses in females exposed to behavioral tests after various endocrine treatments. T-treated females displayed RCSM, but never copulated when exposed to another female. Accordingly they showed an increased Fos expression in POM after ASB but not CSB tests. Females treated with the aromatase inhibitor Vorozole in ovo and T in adulthood displayed both male-typical ASB and CSB, and Fos expression in POM was increased after both types of tests. Thus, the neural circuit mediating ASB is present or can develop in both sexes, but is inactive in females unless they are exposed to exogenous T. In contrast, the neural mechanism mediating CSB is not normally present in females, but can be preserved by blocking the embryonic production of estrogens. Overall these data confirm the difference in endocrine controls and probably neural mechanisms supporting ASB and CSB in quail, and highlight the complexity of mechanisms underlying sexual differentiation of behavior.
Collapse
Affiliation(s)
- Jacques Balthazart
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium.
| | | | | | | |
Collapse
|
50
|
Suárez R, García-González D, de Castro F. Mutual influences between the main olfactory and vomeronasal systems in development and evolution. Front Neuroanat 2012; 6:50. [PMID: 23269914 PMCID: PMC3529325 DOI: 10.3389/fnana.2012.00050] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 11/26/2012] [Indexed: 12/14/2022] Open
Abstract
The sense of smell plays a crucial role in the sensory world of animals. Two chemosensory systems have been traditionally thought to play-independent roles in mammalian olfaction. According to this, the main olfactory system (MOS) specializes in the detection of environmental odorants, while the vomeronasal system (VNS) senses pheromones and semiochemicals produced by individuals of the same or different species. Although both systems differ in their anatomy and function, recent evidence suggests they act synergistically in the perception of scents. These interactions include similar responses to some ligands, overlap of telencephalic connections and mutual influences in the regulation of olfactory-guided behavior. In the present work, we propose the idea that the relationships between systems observed at the organismic level result from a constant interaction during development and reflects a common history of ecological adaptations in evolution. We review the literature to illustrate examples of developmental and evolutionary processes that evidence these interactions and propose that future research integrating both systems may shed new light on the mechanisms of olfaction.
Collapse
Affiliation(s)
- Rodrigo Suárez
- Queensland Brain Institute, The University of Queensland, St Lucia Brisbane, QLD, Australia ; Departamento de Biología, Facultad de Ciencias, Universidad de Chile Santiago, Chile
| | | | | |
Collapse
|