51
|
Cabrera Zapata LE, Garcia-Segura LM, Cambiasso MJ, Arevalo MA. Genetics and Epigenetics of the X and Y Chromosomes in the Sexual Differentiation of the Brain. Int J Mol Sci 2022; 23:ijms232012288. [PMID: 36293143 PMCID: PMC9603441 DOI: 10.3390/ijms232012288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.
Collapse
Affiliation(s)
- Lucas E. Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| | | | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Cátedra de Biología Celular, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Correspondence: (M.J.C.); (M.A.A.)
| | - Maria Angeles Arevalo
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (M.J.C.); (M.A.A.)
| |
Collapse
|
52
|
Beltz AM. Hormonal contraceptive influences on cognition and psychopathology: Past methods, present inferences, and future directions. Front Neuroendocrinol 2022; 67:101037. [PMID: 36154817 DOI: 10.1016/j.yfrne.2022.101037] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/24/2022] [Accepted: 09/16/2022] [Indexed: 11/04/2022]
Abstract
In the last decade, there has been a remarkable surge in research on the neural and behavioral correlates of hormonal contraceptive use, particularly oral contraceptive use. Questions have evolved swiftly and notably, with studies no longer revealing if hormonal contraceptives matter for the brain and behavior, but rather how, when, and for whom they matter most. Paralleling this shift, the goal of this review is to move beyond an average synthesis of hormonal contraceptive influences on human cognition and psychopathology (and their neural substrates) in order to consider the nature and specificity of effects. Accompanied by an evaluation of study methods and informed by findings from animal models, this consideration uncovers promising areas of research in the next ten years, including potential activational and organizational effects of hormonal contraceptive use, individual differences in effects that matter for the wellbeing of unique individuals, and correlates of intrauterine device use.
Collapse
Affiliation(s)
- Adriene M Beltz
- University of Michigan, 2227 East Hall, 530 Church Street, Ann Abor, MI 48109, USA.
| |
Collapse
|
53
|
Solomon-Lane TK, Butler RM, Hofmann HA. Vasopressin mediates nonapeptide and glucocorticoid signaling and social dynamics in juvenile dominance hierarchies of a highly social cichlid fish. Horm Behav 2022; 145:105238. [PMID: 35932752 DOI: 10.1016/j.yhbeh.2022.105238] [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: 03/16/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022]
Abstract
Early-life social experience can strongly affect adult behavior, yet the behavioral mechanisms underlying developmental trajectories are poorly understood. Here, we use the highly social cichlid, Burton's Mouthbrooder (Astatotilapia burtoni) to investigate juvenile social status and behavior, as well as the underlying neuroendocrine mechanisms. We placed juveniles in pairs or triads and found that they readily establish social status hierarchies, with some group structural variation depending on group size, as well as the relative body size of the group members. Next, we used intracerebroventricular injections to test the hypothesis that arginine vasopressin (AVP) regulates juvenile social behavior and status, similar to adult A. burtoni. While we found no direct behavioral effects of experimentally increasing (via vasotocin) or decreasing (via antagonist Manning Compound) AVP signaling, social interactions directed at the treated individual were significantly altered. This group-level effect of central AVP manipulation was also reflected in a significant shift in whole brain expression of genes involved in nonapeptide signaling (AVP, oxytocin, and oxytocin receptor) and the neuroendocrine stress axis (corticotropin-releasing factor (CRF), glucocorticoid receptors (GR) 1a and 1b). Further, social status was associated with the expression of genes involved in glucocorticoid signaling (GR1a, GR1b, GR2, mineralocorticoid receptor), social interactions with the dominant fish, and nonapeptide signaling activity (AVP, AVP receptor V1aR2, OTR). Together, our results considerably expand our understanding of the context-specific emergence of social dominance hierarchies in juveniles and demonstrate a role for nonapeptide and stress axis signaling in the regulation of social status and social group dynamics.
Collapse
Affiliation(s)
- Tessa K Solomon-Lane
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States of America; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States of America.
| | - Rebecca M Butler
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Hans A Hofmann
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States of America; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, United States of America; Institute for Cell & Molecular Biology, The University of Texas at Austin, Austin, TX 78712, United States of America
| |
Collapse
|
54
|
Abstract
Understanding sex differences in physiology and disease requires the identification of the molecular agents that cause phenotypic sex differences. Two groups of such agents are genes located on the sex chromosomes, and gonadal hormones. The former have coherent linkage to chromosomes that form differently in the two sexes under the influence of genomic forces that are not related to reproductive function, whereas the latter have a direct or indirect relationship to reproduction. Evidence published in the past 5 years supports the identification of several agents of sexual differentiation encoded by the X chromosome in mice, including Kdm5c, Kdm6a, Ogt and Xist. These X chromosome agents have wide pleiotropic effects, potentially influencing sex differences in many different tissues, a characteristic shared with the gonadal hormones. The identification of X chromosome agents of sexual differentiation will facilitate understanding of complex intersecting gene pathways underlying sex differences in disease.
Collapse
Affiliation(s)
- Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
55
|
Chen YW, Canli T. "Nothing to see here": No structural brain differences as a function of the Big Five personality traits from a systematic review and meta-analysis. PERSONALITY NEUROSCIENCE 2022; 5:e8. [PMID: 35991756 PMCID: PMC9379932 DOI: 10.1017/pen.2021.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/03/2021] [Accepted: 10/20/2021] [Indexed: 11/24/2022]
Abstract
Personality reflects social, affective, and cognitive predispositions that emerge from genetic and environmental influences. Contemporary personality theories conceptualize a Big Five Model of personality based on the traits of neuroticism, extraversion, agreeableness, conscientiousness, and openness to experience. Starting around the turn of the millennium, neuroimaging studies began to investigate functional and structural brain features associated with these traits. Here, we present the first study to systematically evaluate the entire published literature of the association between the Big Five traits and three different measures of brain structure. Qualitative results were highly heterogeneous, and a quantitative meta-analysis did not produce any replicable results. The present study provides a comprehensive evaluation of the literature and its limitations, including sample heterogeneity, Big Five personality instruments, structural image data acquisition, processing, and analytic strategies, and the heterogeneous nature of personality and brain structures. We propose to rethink the biological basis of personality traits and identify ways in which the field of personality neuroscience can be strengthened in its methodological rigor and replicability.
Collapse
Affiliation(s)
- Yen-Wen Chen
- Program in Integrative Neuroscience, Department of Psychology, Stony Brook University, Stony Brook, NY, USA
| | - Turhan Canli
- Program in Integrative Neuroscience, Department of Psychology, Stony Brook University, Stony Brook, NY, USA
- Department of Psychiatry, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
56
|
Friedrich SR, Nevue AA, Andrade ALP, Velho TAF, Mello CV. Emergence of sex-specific transcriptomes in a sexually dimorphic brain nucleus. Cell Rep 2022; 40:111152. [PMID: 35926465 PMCID: PMC9385264 DOI: 10.1016/j.celrep.2022.111152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 04/26/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
We present the transcriptomic changes underlying the development of an extreme neuroanatomical sex difference. The robust nucleus of the arcopallium (RA) is a key component of the songbird vocal motor system. In zebra finch, the RA is initially monomorphic and then atrophies in females but grows up to 7-fold larger in males. Mirroring this divergence, we show here that sex-differential gene expression in the RA expands from hundreds of predominantly sex chromosome Z genes in early development to thousands of predominantly autosomal genes by the time sexual dimorphism asymptotes. Male-specific developmental processes include cell and axonal growth, synapse assembly and activity, and energy metabolism; female-specific processes include cell polarity and differentiation, transcriptional repression, and steroid hormone and immune signaling. Transcription factor binding site analyses support female-biased activation of pro-apoptotic regulatory networks. The extensive and sex-specific transcriptomic reorganization of RA provides insights into potential drivers of sexually dimorphic neurodevelopment.
Collapse
Affiliation(s)
- Samantha R Friedrich
- Department of Behavioral Neuroscience, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Alexander A Nevue
- Department of Behavioral Neuroscience, Oregon Health & Science University (OHSU), Portland, OR 97239, USA
| | - Abraão L P Andrade
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59078-970, Brazil
| | - Tarciso A F Velho
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN 59078-970, Brazil
| | - Claudio V Mello
- Department of Behavioral Neuroscience, Oregon Health & Science University (OHSU), Portland, OR 97239, USA.
| |
Collapse
|
57
|
Sex differences in the human brain: a roadmap for more careful analysis and interpretation of a biological reality. Biol Sex Differ 2022; 13:43. [PMID: 35883159 PMCID: PMC9327177 DOI: 10.1186/s13293-022-00448-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
The presence, magnitude, and significance of sex differences in the human brain are hotly debated topics in the scientific community and popular media. This debate is largely fueled by studies containing strong, opposing conclusions: either little to no evidence exists for sex differences in human neuroanatomy, or there are small-to-moderate differences in the size of certain brain regions that are highly reproducible across cohorts (even after controlling for sex differences in average brain size). Our Commentary uses the specific comparison between two recent large-scale studies that adopt these opposing views-namely the review by Eliot and colleagues (2021) and the direct analysis of ~ 40k brains by Williams and colleagues (2021)-in an effort to clarify this controversy and provide a framework for conducting this research. First, we review observations that motivate research on sex differences in human neuroanatomy, including potential causes (evolutionary, genetic, and environmental) and effects (epidemiological and clinical evidence for sex-biased brain disorders). We also summarize methodological and empirical support for using structural MRI to investigate such patterns. Next, we outline how researchers focused on sex differences can better specify their study design (e.g., how sex was defined, if and how brain size was adjusted for) and results (by e.g., distinguishing sexual dimorphisms from sex differences). We then compare the different approaches available for studying sex differences across a large number of individuals: direct analysis, meta-analysis, and review. We stress that reviews do not account for methodological differences across studies, and that this variation explains many of the apparent inconsistencies reported throughout recent reviews (including the work by Eliot and colleagues). For instance, we show that amygdala volume is consistently reported as male-biased in studies with sufficient sample sizes and appropriate methods for brain size correction. In fact, comparing the results from multiple large direct analyses highlights small, highly reproducible sex differences in the volume of many brain regions (controlling for brain size). Finally, we describe best practices for the presentation and interpretation of these findings. Care in interpretation is important for all domains of science, but especially so for research on sex differences in the human brain, given the existence of broad societal gender-biases and a history of biological data being used justify sexist ideas. As such, we urge researchers to discuss their results from simultaneously scientific and anti-sexist viewpoints.
Collapse
|
58
|
Kelly AM, Gonzalez Abreu JA, Thompson RR. Beyond sex and aggression: testosterone rapidly matches behavioural responses to social context and tries to predict the future. Proc Biol Sci 2022; 289:20220453. [PMID: 35673866 PMCID: PMC9174716 DOI: 10.1098/rspb.2022.0453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Although androgens are widely studied in the context of aggression, androgenic influences on prosocial behaviours have been less explored. We examined testosterone's (T) influence on prosocial and aggressive responses in a positively valenced social context (interacting with a pairbond partner) and a negatively valenced context (interacting with an intruder) in socially monogamous Mongolian gerbils. T increased and decreased prosocial responses in the same individuals towards a pairbond partner and an intruder, respectively, both within 30 min, but did not affect aggression. T also had persistent effects on prosocial behaviour; males in which T initially increased prosocial responses towards a partner continued to exhibit elevated prosocial responses towards an intruder male days later until a second T injection rapidly eliminated those responses. Thus, T surges can rapidly match behaviour to current social context, as well as prime animals for positive social interactions in the future. Neuroanatomically, T rapidly increased hypothalamic oxytocin, but not vasopressin, cellular responses during interactions with a partner. Together, our results indicate that T can facilitate and inhibit prosocial behaviours depending on social context, that it can influence prosocial responses across rapid and prolonged time scales, and that it affects oxytocin signalling mechanisms that could mediate its context-dependent behavioural influences.
Collapse
Affiliation(s)
- Aubrey M. Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | | | - Richmond R. Thompson
- Division of Social Sciences, Oxford College of Emory University, 801 Emory Street, Oxford GA 30054 USA
| |
Collapse
|
59
|
Arnold AP. Integrating Sex Chromosome and Endocrine Theories to Improve Teaching of Sexual Differentiation. Cold Spring Harb Perspect Biol 2022; 14:a039057. [PMID: 35667790 PMCID: PMC9438782 DOI: 10.1101/cshperspect.a039057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Major sex differences in mammalian tissues are functionally tied to reproduction and evolved as adaptations to meet different reproductive needs of females and males. They were thus directly controlled by gonadal hormones. Factors encoded on the sex chromosomes also cause many sex differences in diverse tissues because they are present in different doses in XX and XY cells. The sex chromosome effects likely evolved not because of demands of reproduction, but as side effects of genomic forces that adaptively reduced sexual inequality. Sex-specific effects of particular factors, including gonadal hormones, therefore, are not necessarily explained as adaptations for reproduction, but also as potential factors offsetting, rather than producing, sex differences. The incorporation of these concepts would improve future teaching about sexual differentiation.
Collapse
Affiliation(s)
- Arthur P Arnold
- Department of Integrative Biology & Physiology, and Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, Los Angeles, California 90095-7239, USA
| |
Collapse
|
60
|
Gonzalez Abreu JA, Rosenberg AE, Fricker BA, Wallace KJ, Seifert AW, Kelly AM. Species-typical group size differentially influences social reward neural circuitry during nonreproductive social interactions. iScience 2022; 25:104230. [PMID: 35521530 PMCID: PMC9062245 DOI: 10.1016/j.isci.2022.104230] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/24/2022] [Accepted: 04/06/2022] [Indexed: 11/22/2022] Open
Abstract
We investigated whether nonreproductive social interactions may be rewarding for colonial but not non-colonial species. We found that the colonial spiny mouse (Acomys cahirinus) is significantly more gregarious, more prosocial, and less aggressive than its non-colonial relative, the Mongolian gerbil (Meriones unguiculatus). In an immediate-early gene study, we examined oxytocin (OT) and tyrosine hydroxylase (TH) neural responses to interactions with a novel, same-sex conspecific or a novel object. The paraventricular nucleus of the hypothalamus (PVN) OT cell group was more responsive to interactions with a conspecific compared to a novel object in both species. However, the ventral tegmental area (VTA) TH cell group showed differential responses only in spiny mice. Further, PVN OT and VTA TH neural responses positively correlated in spiny mice, suggesting functional connectivity. These results suggest that colonial species may have evolved neural mechanisms associated with reward in novel, nonreproductive social contexts to promote large group-living.
Collapse
Affiliation(s)
| | - Ashley E. Rosenberg
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Brandon A. Fricker
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Kelly J. Wallace
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, 675 Rose Street, Lexington, KY 40506, USA
| | - Aubrey M. Kelly
- Department of Psychology, Emory University, 36 Eagle Row, Atlanta, GA 30322, USA
| |
Collapse
|
61
|
du Plessis KC, Basu S, Rumbell TH, Lucas EK. Sex-Specific Neural Networks of Cued Threat Conditioning: A Pilot Study. Front Syst Neurosci 2022; 16:832484. [PMID: 35656357 PMCID: PMC9152023 DOI: 10.3389/fnsys.2022.832484] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 04/07/2022] [Indexed: 11/28/2022] Open
Abstract
Cued threat conditioning is the most common preclinical model for emotional memory, which is dysregulated in anxiety disorders and post-traumatic stress disorder. Though women are twice as likely as men to develop these disorders, current knowledge of threat conditioning networks was established by studies that excluded female subjects. For unbiased investigation of sex differences in these networks, we quantified the neural activity marker c-fos across 112 brain regions in adult male and female mice after cued threat conditioning compared to naïve controls. We found that trained females engaged prelimbic cortex, lateral amygdala, cortical amygdala, dorsal peduncular cortex, and subparafasicular nucleus more than, and subparaventricular zone less than, trained males. To explore how these sex differences in regional activity impact the global network, we generated interregional cross-correlations of c-fos expression to identify regions that were co-active during conditioning and performed hub analyses to identify regional control centers within each neural network. These exploratory graph theory-derived analyses revealed sex differences in the functional coordination of the threat conditioning network as well as distinct hub regions between trained males and females. Hub identification across multiple networks constructed by sequentially pruning the least reliable connections revealed globus pallidus and ventral lateral septum as the most robust hubs for trained males and females, respectively. While low sample size and lack of non-associative controls are major limitations, these findings provide preliminary evidence of sex differences in the individual circuit components and broader global networks of threat conditioning that may confer female vulnerability to fear-based psychiatric disease.
Collapse
Affiliation(s)
- Kamryn C. du Plessis
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sreetama Basu
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, United States
| | - Timothy H. Rumbell
- IBM Thomas J. Watson Research Center, Yorktown Heights, NY, United States
| | - Elizabeth K. Lucas
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
- *Correspondence: Elizabeth K. Lucas,
| |
Collapse
|
62
|
Spool JA, Bergan JF, Remage-Healey L. A neural circuit perspective on brain aromatase. Front Neuroendocrinol 2022; 65:100973. [PMID: 34942232 PMCID: PMC9667830 DOI: 10.1016/j.yfrne.2021.100973] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
This review explores the role of aromatase in the brain as illuminated by a set of conserved network-level connections identified in several vertebrate taxa. Aromatase-expressing neurons are neurochemically heterogeneous but the brain regions in which they are found are highly-conserved across the vertebrate lineage. During development, aromatase neurons have a prominent role in sexual differentiation of the brain and resultant sex differences in behavior and human brain diseases. Drawing on literature primarily from birds and rodents, we delineate brain regions that express aromatase and that are strongly interconnected, and suggest that, in many species, aromatase expression essentially defines the Social Behavior Network. Moreover, in several cases the inputs to and outputs from this core Social Behavior Network also express aromatase. Recent advances in molecular and genetic tools for neuroscience now enable in-depth and taxonomically diverse studies of the function of aromatase at the neural circuit level.
Collapse
Affiliation(s)
- Jeremy A Spool
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Joseph F Bergan
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Luke Remage-Healey
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
63
|
Estrogenic Action in Stress-Induced Neuroendocrine Regulation of Energy Homeostasis. Cells 2022; 11:cells11050879. [PMID: 35269500 PMCID: PMC8909319 DOI: 10.3390/cells11050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
Abstract
Estrogens are among important contributing factors to many sex differences in neuroendocrine regulation of energy homeostasis induced by stress. Research in this field is warranted since chronic stress-related psychiatric and metabolic disturbances continue to be top health concerns, and sex differences are witnessed in these aspects. For example, chronic stress disrupts energy homeostasis, leading to negative consequences in the regulation of emotion and metabolism. Females are known to be more vulnerable to the psychological consequences of stress, such as depression and anxiety, whereas males are more vulnerable to the metabolic consequences of stress. Sex differences that exist in the susceptibility to various stress-induced disorders have led researchers to hypothesize that gonadal hormones are regulatory factors that should be considered in stress studies. Further, estrogens are heavily recognized for their protective effects on metabolic dysregulation, such as anti-obesogenic and glucose-sensing effects. Perturbations to energy homeostasis using laboratory rodents, such as physiological stress or over-/under- feeding dietary regimen prevalent in today’s society, offer hints to the underlying mechanisms of estrogenic actions. Metabolic effects of estrogens primarily work through estrogen receptor α (ERα), which is differentially expressed between the sexes in hypothalamic nuclei regulating energy metabolism and in extrahypothalamic limbic regions that are not typically associated with energy homeostasis. In this review, we discuss estrogenic actions implicated in stress-induced sex-distinct metabolic disorders.
Collapse
|
64
|
Vahaba DM, Halstead ER, Donaldson ZR, Ahern TH, Beery AK. Sex differences in the reward value of familiar mates in prairie voles. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12790. [PMID: 35044087 PMCID: PMC8917082 DOI: 10.1111/gbb.12790] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/17/2022]
Abstract
The rewarding properties of social interactions facilitate relationship formation and maintenance. Prairie voles are one of the few laboratory species that form selective relationships, manifested as "partner preferences" for familiar partners versus strangers. While both sexes exhibit strong partner preferences, this similarity in outward behavior likely results from sex-specific neurobiological mechanisms. We recently demonstrated that in operant trials, females worked hardest for access to familiar conspecifics of either sex, while males worked equally hard for access to any female, indicating a sex difference in social motivation. As tests were performed with one social target at a time, males might have experienced a ceiling effect, and familiar females might be more relatively rewarding in a choice scenario. Here we performed an operant social choice task in which voles lever-pressed to gain temporary access to either the chamber containing their mate or one containing a novel opposite-sex vole. Females worked hardest to access their mate, while males pressed at similar rates for either female. Individual male behavior was heterogeneous, congruent with multiple mating strategies in the wild. Voles exhibited preferences for favorable over unfavorable environments in a non-social operant task, indicating that lack of social preference does not reflect lack of discrimination. Natural variation in oxytocin receptor genotype at the intronic single nucleotide polymorphism NT213739 was associated with oxytocin receptor density, and predicted individual variation in stranger-directed aggressive behavior. These findings suggest that convergent preference behavior in male and female voles results from sex-divergent pathways, particularly in the realm of social motivation.
Collapse
Affiliation(s)
- Daniel M. Vahaba
- Program in Neuroscience, Department of BiologySmith CollegeNorthamptonMassachusettsUSA
| | - Emily R. Halstead
- Program in Neuroscience, Department of BiologySmith CollegeNorthamptonMassachusettsUSA
| | - Zoe R. Donaldson
- Department of Molecular, Cellular, and Developmental Biology, Department of Psychology & NeuroscienceUniversity of Colorado BoulderBoulderColoradoUSA
| | - Todd H. Ahern
- Center for Behavioral NeuroscienceQuinnipiac UniversityHamdenConnecticutUSA
| | - Annaliese K. Beery
- Program in Neuroscience, Department of BiologySmith CollegeNorthamptonMassachusettsUSA,Department of Integrative BiologyUniversity of California BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|
65
|
Rochat MJ. Sex and gender differences in the development of empathy. J Neurosci Res 2022; 101:718-729. [PMID: 35043464 DOI: 10.1002/jnr.25009] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 01/10/2023]
Abstract
The topic of typical sex and gender difference in empathy is examined in both a developmental and neuroscientific perspective. Empathy is construed as a multi-layered phenomenon with various degrees of complexity unfolding in ontogeny. The different components of empathy (i.e., affective, cognitive, and prosocial motivation) will be discussed as they interact and are expressed behaviorally. Significant sex/gender differences in empathy are discussed in relation to putative bottom-up or top-down processes underlying empathetic responses. The early onset and the pervasive presence of such sex/gender differences throughout the lifespan are further discussed in light of social and neurobiological modeling factors, including early socialization, brain's structural/functional variances, as well as genetics and hormonal factors.
Collapse
Affiliation(s)
- Magali Jane Rochat
- Functional and Molecular Neuroimaging Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| |
Collapse
|
66
|
Parel ST, Peña CJ. Genome-wide Signatures of Early-Life Stress: Influence of Sex. Biol Psychiatry 2022; 91:36-42. [PMID: 33602500 PMCID: PMC8791071 DOI: 10.1016/j.biopsych.2020.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 01/03/2023]
Abstract
Both history of early-life stress (ELS) and female sex are associated with increased risk for depression. The complexity of how ELS interacts with brain development and sex to impart risk for multifaceted neuropsychiatric disorders is also unlikely to be understood by examining changes in single genes. Here, we review an emerging literature on genome-wide transcriptional and epigenetic signatures of ELS and the potential moderating influence of sex. We discuss evidence both that there are latent sex differences revealed by ELS and that ELS itself produces latent transcriptomic changes revealed by adult stress. In instances where there are broad similarities in global signatures of ELS among females and males, genes that contribute to these patterns are largely distinct based on sex. As this area of investigation grows, an effort should be made to better understand the sex-specific impact of ELS within the human brain, specific contributions of chromosomal versus hormonal sex, how ELS alters the time course of normal transcriptional development, and the cell-type specificity of transcriptomic and epigenomic changes in the brain. A better understanding of how ELS interacts with sex to alter transcriptomic and epigenomic signatures in the brain will inform individualized therapeutic strategies to prevent or ameliorate depression and other psychiatric disorders in this vulnerable population.
Collapse
Affiliation(s)
- Sero Toriano Parel
- Princeton Neuroscience Institute, Princeton University, Princeton, New Jersey
| | | |
Collapse
|
67
|
Farrar VS, Harris RM, Austin SH, Nava Ultreras BM, Booth AM, Angelier F, Lang AS, Feustel T, Lee C, Bond A, MacManes MD, Calisi RM. Prolactin and prolactin receptor expression in the HPG axis and crop during parental care in both sexes of a biparental bird (Columba livia). Gen Comp Endocrinol 2022; 315:113940. [PMID: 34756919 DOI: 10.1016/j.ygcen.2021.113940] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/11/2021] [Accepted: 10/22/2021] [Indexed: 12/31/2022]
Abstract
During breeding, multiple circulating hormones, including prolactin, facilitate reproductive transitions in species that exhibit parental care. Prolactin underlies parental behaviors and related physiological changes across many vertebrates, including birds and mammals. While circulating prolactin levels often fluctuate across breeding, less is known about how relevant target tissues vary in their prolactin responsiveness via prolactin receptor (PRLR) expression. Recent studies have also investigated prolactin (PRL) gene expression outside of the pituitary (i.e., extra-pituitary PRL), but how PRL gene expression varies during parental care in non-pituitary tissue (e.g., hypothalamus, gonads) remains largely unknown. Further, it is unclear if and how tissue-specific PRL and PRLR vary between the sexes during biparental care. To address this, we measured PRL and PRLR gene expression in tissues relevant to parental care, the endocrine reproductive hypothalamic-pituitary- gonadal (HPG) axis and the crop (a tissue with a similar function as the mammalian mammary gland), across various reproductive stages in both sexes of a biparental bird, the rock dove (Columba livia). We also assessed how these genes responded to changes in offspring presence by adding chicks mid-incubation, simulating an early hatch when prolactin levels were still moderately low. We found that pituitary PRL expression showed similar increases as plasma prolactin levels, and detected extra-pituitary PRL in the hypothalamus, gonads and crop. Hypothalamic and gonadal PRLR expression also changed as birds began incubation. Crop PRLR expression correlated with plasma prolactin, peaking when chicks hatched. In response to replacing eggs with a novel chick mid-incubation, hypothalamic and gonadal PRL and PRLR gene expression differed significantly compared to mid-incubation controls, even when plasma prolactin levels did not differ. We also found sex differences in PRL and PRLR that suggest gene expression may allow males to compensate for lower levels in prolactin by upregulating PRLR in all tissues. Overall, this study advances our understanding of how tissue-specific changes in responsiveness to parental hormones may differ across key reproductive transitions, in response to offspring cues, and between the sexes.
Collapse
Affiliation(s)
- Victoria S Farrar
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States.
| | - Rayna M Harris
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Suzanne H Austin
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Brandon M Nava Ultreras
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - April M Booth
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Frédéric Angelier
- Centre d'Etudes Biologiques de Chizé, CNRS, UMR 7372, 79360 Villiers en Bois, France
| | - Andrew S Lang
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, United States
| | - Tanner Feustel
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Candice Lee
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Annie Bond
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| | - Matthew D MacManes
- Department of Molecular, Cellular and Biomedical Sciences, University of New Hampshire, Durham, NH 03824, United States
| | - Rebecca M Calisi
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, Davis, CA 95616, United States
| |
Collapse
|
68
|
Long-lasting Postnatal Sensory Deprivation Alters Dendritic Morphology of Pyramidal Neurons in the Rat Hippocampus: Behavioral Correlates. Neuroscience 2022; 480:79-96. [PMID: 34785272 DOI: 10.1016/j.neuroscience.2021.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/20/2022]
Abstract
The role of normal sensory inputs in the development of sensory cortices is well known, however, their impacts on the hippocampus, an integrator of sensory modalities with important roles in cognitive functions, has received much less attention. Here, we applied a long-term sensory deprivation paradigm by trimming the rats' whiskers bilaterally, from postnatal day 3 to 59. Female sensory-deprived (SD) rats showed more on-wall rearing and visits to the center of the open-field box, shorter periods of grooming, less defecation and less anxiety-like behaviors in the elevated plus-maze compared to controls, who had their intact whiskers brushed. Passive avoidance memory retention was sex-dependently impaired in the female SD rats. In the radial arm maze, however, reference spatial memory was impaired only in the male SD rats. Nonetheless, working memory errors increased in both sexes of SD rats. Besides depletion of CA1 and CA3 pyramidal neurons in SD rats, Sholl analysis of Golgi-Cox stained neurons revealed that prolonged sensory deprivation has retracted the arborization of CA1 basal dendrites in SD group, while solely female SD rats had diminished CA1 apical dendrites. Sholl analysis of CA3 neurons in SD animals also disclosed significantly more branched apical dendrites in males and basal dendrites in females. Sensory deprivation also led to a considerable spine loss and variation of different spine types in a sex-dependent manner. Our findings suggest that experience-dependent structural plasticity is capable of spreading far beyond the manipulated sensory zones and the inevitable functional alterations can be expressed in a multifactorial sex-dependent manner.
Collapse
|
69
|
Thorne BN, Ellenbroek BA, Day DJ. Sex bias in the serotonin transporter knockout model: implications for neuropsychiatric disorder research. Neurosci Biobehav Rev 2022; 134:104547. [DOI: 10.1016/j.neubiorev.2022.104547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/09/2022] [Accepted: 01/18/2022] [Indexed: 12/22/2022]
|
70
|
Seney ML, Glausier J, Sibille E. Large-Scale Transcriptomics Studies Provide Insight Into Sex Differences in Depression. Biol Psychiatry 2022; 91:14-24. [PMID: 33648716 PMCID: PMC8263802 DOI: 10.1016/j.biopsych.2020.12.025] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/11/2022]
Abstract
Major depressive disorder (MDD) is a leading cause of disability, affecting more than 300 million people worldwide. We first review the well-known sex difference in incidence of MDD, with women being twice as likely to be diagnosed as men, and briefly summarize how the impact of MDD varies between men and women, with sex differences in symptoms, severity, and antidepressant drug response. We then attempt to deconstruct the biological bases for MDD and discuss implications for sex differences research. Next, we review findings from human postmortem studies, both from selected candidate gene studies and from well-powered, unbiased transcriptomics studies, which suggest distinct, and possibly opposite, molecular changes in the brains of depressed men and women. We then discuss inherent challenges of research on the human postmortem brain and suggest paths forward that rely on thoughtful cohort design. Although studies indicate that circulating gonadal hormones might underlie the observed sex differences in MDD, we discuss how additional sex-specific factors, such as genetic sex and developmental exposure to gonadal hormones, may also contribute to altered vulnerability, and we highlight various nuances that we believe should be considered when determining mechanisms underlying observed sex differences. Altogether, this review highlights not only how various sex-specific factors might influence susceptibility or resilience to depression, but also how those sex-specific factors might result in divergent pathology in men and women.
Collapse
Affiliation(s)
- Marianne L Seney
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania.
| | - Jill Glausier
- Department of Psychiatry, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania; Translational Neuroscience Program, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
71
|
Everts R, Muri R, Leibundgut K, Siegwart V, Wiest R, Steinlin M. Fear and discomfort of children and adolescents during MRI: ethical consideration on research MRIs in children. Pediatr Res 2022; 91:720-723. [PMID: 33879848 PMCID: PMC9064788 DOI: 10.1038/s41390-020-01277-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Regula Everts
- Division of Neuropediatrics, Development and Rehabilitation, Children's University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland. .,Department of Pediatric Hematology and Oncology, Children's University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Raphaela Muri
- grid.411656.10000 0004 0479 0855Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland ,grid.411656.10000 0004 0479 0855Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kurt Leibundgut
- grid.411656.10000 0004 0479 0855Department of Pediatric Hematology and Oncology, Children’s University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Valerie Siegwart
- grid.411656.10000 0004 0479 0855Division of Neuropediatrics, Development and Rehabilitation, Children’s University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland ,grid.411656.10000 0004 0479 0855Department of Pediatric Hematology and Oncology, Children’s University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Roland Wiest
- grid.411656.10000 0004 0479 0855Institute of Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Maja Steinlin
- grid.411656.10000 0004 0479 0855Division of Neuropediatrics, Development and Rehabilitation, Children’s University Hospital, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
72
|
Landen S, Jacques M, Hiam D, Alvarez-Romero J, Harvey NR, Haupt LM, Griffiths LR, Ashton KJ, Lamon S, Voisin S, Eynon N. Skeletal muscle methylome and transcriptome integration reveals profound sex differences related to muscle function and substrate metabolism. Clin Epigenetics 2021; 13:202. [PMID: 34732242 PMCID: PMC8567658 DOI: 10.1186/s13148-021-01188-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/19/2021] [Indexed: 12/29/2022] Open
Abstract
Nearly all human complex traits and diseases exhibit some degree of sex differences, with epigenetics being one of the main contributing factors. Various tissues display sex differences in DNA methylation; however, this has not yet been explored in skeletal muscle, despite skeletal muscle being among the tissues with the most transcriptomic sex differences. For the first time, we investigated the effect of sex on autosomal DNA methylation in human skeletal muscle across three independent cohorts (Gene SMART, FUSION, and GSE38291) using a meta-analysis approach, totalling 369 human muscle samples (222 males and 147 females), and integrated this with known sex-biased transcriptomics. We found 10,240 differentially methylated regions (DMRs) at FDR < 0.005, 94% of which were hypomethylated in males, and gene set enrichment analysis revealed that differentially methylated genes were involved in muscle contraction and substrate metabolism. We then investigated biological factors underlying DNA methylation sex differences and found that circulating hormones were not associated with differential methylation at sex-biased DNA methylation loci; however, these sex-specific loci were enriched for binding sites of hormone-related transcription factors (with top TFs including androgen (AR), estrogen (ESR1), and glucocorticoid (NR3C1) receptors). Fibre type proportions were associated with differential methylation across the genome, as well as across 16% of sex-biased DNA methylation loci (FDR < 0.005). Integration of DNA methylomic results with transcriptomic data from the GTEx database and the FUSION cohort revealed 326 autosomal genes that display sex differences at both the epigenome and transcriptome levels. Importantly, transcriptional sex-biased genes were overrepresented among epigenetic sex-biased genes (p value = 4.6e−13), suggesting differential DNA methylation and gene expression between male and female muscle are functionally linked. Finally, we validated expression of three genes with large effect sizes (FOXO3A, ALDH1A1, and GGT7) in the Gene SMART cohort with qPCR. GGT7, involved in antioxidant metabolism, displays male-biased expression as well as lower methylation in males across the three cohorts. In conclusion, we uncovered 8420 genes that exhibit DNA methylation differences between males and females in human skeletal muscle that may modulate mechanisms controlling muscle metabolism and health.
Collapse
Affiliation(s)
- Shanie Landen
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia
| | - Macsue Jacques
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia
| | - Danielle Hiam
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia.,Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Javier Alvarez-Romero
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia
| | - Nicholas R Harvey
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, 4226, Australia.,Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD, 4059, Australia
| | - Larisa M Haupt
- Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD, 4059, Australia
| | - Lyn R Griffiths
- Centre for Genomics and Personalised Health, Genomics Research Centre, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD, 4059, Australia
| | - Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, 4226, Australia
| | - Séverine Lamon
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Sarah Voisin
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia
| | - Nir Eynon
- Institute for Health and Sport (iHeS), Victoria University, PO Box 14428, Melbourne, VIC, 8001, Australia.
| |
Collapse
|
73
|
Beery AK, Lopez SA, Blandino KL, Lee NS, Bourdon NS. Social selectivity and social motivation in voles. eLife 2021; 10:e72684. [PMID: 34726153 PMCID: PMC8594915 DOI: 10.7554/elife.72684] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 10/19/2021] [Indexed: 01/19/2023] Open
Abstract
Selective relationships are fundamental to humans and many other animals, but relationships between mates, family members, or peers may be mediated differently. We examined connections between social reward and social selectivity, aggression, and oxytocin receptor signaling pathways in rodents that naturally form enduring, selective relationships with mates and peers (monogamous prairie voles) or peers (group-living meadow voles). Female prairie and meadow voles worked harder to access familiar versus unfamiliar individuals, regardless of sex, and huddled extensively with familiar subjects. Male prairie voles displayed strongly selective huddling preferences for familiar animals, but only worked harder to repeatedly access females versus males, with no difference in effort by familiarity. This reveals a striking sex difference in pathways underlying social monogamy and demonstrates a fundamental disconnect between motivation and social selectivity in males-a distinction not detected by the partner preference test. Meadow voles exhibited social preferences but low social motivation, consistent with tolerance rather than reward supporting social groups in this species. Natural variation in oxytocin receptor binding predicted individual variation in prosocial and aggressive behaviors. These results provide a basis for understanding species, sex, and individual differences in the mechanisms underlying the role of social reward in social preference.
Collapse
Affiliation(s)
- Annaliese K Beery
- Department of Integrative Biology, University of California BerkeleyBerkeleyUnited States
- Program in Neuroscience, Departments of Psychology and Biology, Smith CollegeNorthamptonUnited States
- Neuroscience and Behavior Graduate Program, University of MassachusettsAmherst, MAUnited States
| | - Sarah A Lopez
- Program in Neuroscience, Departments of Psychology and Biology, Smith CollegeNorthamptonUnited States
| | - Katrina L Blandino
- Program in Neuroscience, Departments of Psychology and Biology, Smith CollegeNorthamptonUnited States
| | - Nicole S Lee
- Neuroscience and Behavior Graduate Program, University of MassachusettsAmherst, MAUnited States
| | - Natalie S Bourdon
- Program in Neuroscience, Departments of Psychology and Biology, Smith CollegeNorthamptonUnited States
| |
Collapse
|
74
|
Sex differences in anxiety and depression: circuits and mechanisms. Nat Rev Neurosci 2021; 22:674-684. [PMID: 34545241 DOI: 10.1038/s41583-021-00513-0] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 02/08/2023]
Abstract
Epidemiological sex differences in anxiety disorders and major depression are well characterized. Yet the circuits and mechanisms that contribute to these differences are understudied, because preclinical studies have historically excluded female rodents. This oversight is beginning to be addressed, and recent studies that include male and female rodents are identifying sex differences in neurobiological processes that underlie features of these disorders, including conflict anxiety, fear processing, arousal, social avoidance, learned helplessness and anhedonia. These findings allow us to conceptualize various types of sex differences in the brain, which in turn have broader implications for considering sex as a biological variable. Importantly, comparing the sexes could aid in the discovery of novel therapeutics.
Collapse
|
75
|
Krawczyk MC, Millan J, Blake MG, Boccia MM. Role of prediction error and the cholinergic system on memory reconsolidation processes in mice. Neurobiol Learn Mem 2021; 185:107534. [PMID: 34619364 DOI: 10.1016/j.nlm.2021.107534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/24/2021] [Accepted: 09/30/2021] [Indexed: 11/26/2022]
Abstract
The ability to make predictions based on stored information is a general coding strategy. A prediction error (PE) is a mismatch between expected and current events. Our memories, like ourselves, are subject to change. Thus, an acquired memory can become active and update its content or strength by a labilization-reconsolidation process. Within the reconsolidation framework, PE drives the updating of consolidated memories. In the past our lab has made key progresses showing that a blockade in the central cholinergic system during reconsolidation can cause memory impairment, while reinforcement of cholinergic activity enhances it. In the present work we determined that PE is a necessary condition for memory to reconsolidate in an inhibitory avoidance task using both male and female mice. Depending on the intensity of the unconditioned stimulus (US) used during training, a negative (higher US intensity) or positive (lower US intensity/no US) PE on a retrieval session modified the behavioral response on a subsequent testing session. Furthermore, we demonstrated that the cholinergic system modulates memory reconsolidation only when PE is detected. In this scenario administration of oxotremorine, scopolamine or nicotine after memory reactivation either enhanced or impaired memory reconsolidation in a sex-specific manner.
Collapse
Affiliation(s)
- M C Krawczyk
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - J Millan
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - M G Blake
- Instituto de Fisiología y Biofísica (IFIBIO UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - M M Boccia
- Laboratorio de Neurofarmacología de los Procesos de Memoria, Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina.
| |
Collapse
|
76
|
Affiliation(s)
- Liisa A M Galea
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, Women's Health Research Cluster, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
77
|
Sex/gender differences in the brain are not trivial-A commentary on Eliot et al. (2021). Neurosci Biobehav Rev 2021; 130:408-409. [PMID: 34509515 DOI: 10.1016/j.neubiorev.2021.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 11/22/2022]
Abstract
In this commentary to the comprehensive review by Eliot et al. (2021), we fully comply with rejecting the 'sexual dimorphism' concept in its extreme, binary form. However, we criticise the authors' extreme position and argue that sex/gender differences in the brain are far from being 'trivial' and 'unlikely to be meaningful'. Our key arguments refer to the importance of small effects which can have meaningful behavioural consequences, and to several non-binary sex/gender-related factors which might explain individual differences better than sex/gender per se and which have shown to play important roles as risk factors in the aetiology of many mental and neurodevelopmental disorders. We conclude that the biopsychosocial approach is key to understanding sex/gender differences in the brain better than we currently do.
Collapse
|
78
|
Yamashita J, Nishiike Y, Fleming T, Kayo D, Okubo K. Estrogen mediates sex differences in preoptic neuropeptide and pituitary hormone production in medaka. Commun Biol 2021; 4:948. [PMID: 34373576 PMCID: PMC8352984 DOI: 10.1038/s42003-021-02476-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/22/2021] [Indexed: 11/21/2022] Open
Abstract
The preoptic area (POA) is one of the most evolutionarily conserved regions of the vertebrate brain and contains subsets of neuropeptide-expressing neurons. Here we found in the teleost medaka that two neuropeptides belonging to the secretin family, pituitary adenylate cyclase-activating polypeptide (Pacap) and vasoactive intestinal peptide (Vip), exhibit opposite patterns of sexually dimorphic expression in the same population of POA neurons that project to the anterior pituitary: Pacap is male-biased, whereas Vip is female-biased. Estrogen secreted by the ovary in adulthood was found to attenuate Pacap expression and, conversely, stimulate Vip expression in the female POA, thereby establishing and maintaining their opposite sexual dimorphism. Pituitary organ culture experiments demonstrated that both Pacap and Vip can markedly alter the expression of various anterior pituitary hormones. Collectively, these findings show that males and females use alternative preoptic neuropeptides to regulate anterior pituitary hormones as a result of their different estrogen milieu.
Collapse
Affiliation(s)
- Junpei Yamashita
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Yuji Nishiike
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Thomas Fleming
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Daichi Kayo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kataaki Okubo
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
79
|
Lenert ME, Avona A, Garner KM, Barron LR, Burton MD. Sensory Neurons, Neuroimmunity, and Pain Modulation by Sex Hormones. Endocrinology 2021; 162:bqab109. [PMID: 34049389 PMCID: PMC8237991 DOI: 10.1210/endocr/bqab109] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Indexed: 12/16/2022]
Abstract
The inclusion of women in preclinical pain studies has become more commonplace in the last decade as the National Institutes of Health (NIH) released its "Sex as a Biological Variable" mandate. Presumably, basic researchers have not had a comprehensive understanding about neuroimmune interactions in half of the population and how hormones play a role in this. To date, we have learned that sex hormones contribute to sexual differentiation of the nervous system and sex differences in behavior throughout the lifespan; however, the cycling of sex hormones does not always explain these differences. Here, we highlight recent advances in our understanding of sex differences and how hormones and immune interactions influence sensory neuron activity to contribute to physiology and pain. Neuroimmune mechanisms may be mediated by different cell types in each sex, as the actions of immune cells are sexually dimorphic. Unfortunately, the majority of studies assessing neuronal contributions to immune function have been limited to males, so it is unclear if the mechanisms are similar in females. Finally, pathways that control cellular metabolism, like nuclear receptors, have been shown to play a regulatory role both in pain and inflammation. Overall, communication between the neuroimmune and endocrine systems modulate pain signaling in a sex-dependent manner, but more research is needed to reveal nuances of these mechanisms.
Collapse
Affiliation(s)
- Melissa E Lenert
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Amanda Avona
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Katherine M Garner
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Luz R Barron
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Center for Advanced Pain Studies (CAPS), Department of Neuroscience, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas 75080, USA
| |
Collapse
|
80
|
Krentzel AA, Kimble LC, Dorris DM, Horman BM, Meitzen J, Patisaul HB. FireMaster® 550 (FM 550) exposure during the perinatal period impacts partner preference behavior and nucleus accumbens core medium spiny neuron electrophysiology in adult male and female prairie voles, Microtus ochrogaster. Horm Behav 2021; 134:105019. [PMID: 34182292 PMCID: PMC8403633 DOI: 10.1016/j.yhbeh.2021.105019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/11/2021] [Indexed: 11/23/2022]
Abstract
One of the most widely used flame retardant (FR) mixtures in household products is Firemaster 550 (FM 550). FM 550 leaches from items such as foam-based furniture and infant products, resulting in contamination of the household environment and biota. Previous studies indicate sex-specific behavioral deficits in rodents and zebrafish in response to developmental FM 550 exposure. These deficits include impacts on social and attachment behaviors in a prosocial rodent: the prairie vole (Microtus ochrogaster). The prairie vole is a laboratory-acclimated rodent that exhibits spontaneous attachment behaviors including pair bonding. Here we extend previous work by addressing how developmental exposure to FM 550 impacts pair bonding strength via an extended-time partner preference test, as well as neuron electrophysiological properties in a region implicated in pair bond behavior, the nucleus accumbens (NAcc) core. Dams were exposed to vehicle or 1000 μg of FM 550 via subcutaneous injections throughout gestation, and female and male pups were directly exposed beginning the day after birth until weaning. Pair bond behavior of adult female and male offspring was assessed using a three hour-long partner preference test. Afterwards, acute brain slices of the NAcc core were produced and medium spiny neuron electrophysiological attributes recorded via whole cell patch-clamp. Behavioral impacts were sex-specific. Partner preference behavior was increased in exposed females but decreased in exposed males. Electrophysiological impacts were similar between sexes and specific to attributes related to input resistance. Input resistance was decreased in neurons recorded from both sexes exposed to FM 550 compared to vehicle. This study supports the hypothesis that developmental exposure to FM 550 impacts attachment behaviors and demonstrates a novel FM 550 effect on neural electrophysiology.
Collapse
Affiliation(s)
- Amanda A Krentzel
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Laney C Kimble
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - David M Dorris
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Brian M Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - John Meitzen
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC 27695, USA.
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
81
|
van Eijk L, Zhu D, Couvy-Duchesne B, Strike LT, Lee AJ, Hansell NK, Thompson PM, de Zubicaray GI, McMahon KL, Wright MJ, Zietsch BP. Are Sex Differences in Human Brain Structure Associated With Sex Differences in Behavior? Psychol Sci 2021; 32:1183-1197. [PMID: 34323639 DOI: 10.1177/0956797621996664] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
On average, men and women differ in brain structure and behavior, raising the possibility of a link between sex differences in brain and behavior. But women and men are also subject to different societal and cultural norms. We navigated this challenge by investigating variability of sex-differentiated brain structure within each sex. Using data from the Queensland Twin IMaging study (n = 1,040) and Human Connectome Project (n = 1,113), we obtained data-driven measures of individual differences along a male-female dimension for brain and behavior based on average sex differences in brain structure and behavior, respectively. We found a weak association between these brain and behavioral differences, driven by brain size. These brain and behavioral differences were moderately heritable. Our findings suggest that behavioral sex differences are, to some extent, related to sex differences in brain structure but that this is mainly driven by differences in brain size, and causality should be interpreted cautiously.
Collapse
Affiliation(s)
- Liza van Eijk
- Centre for Psychology and Evolution, School of Psychology, University of Queensland.,Queensland Brain Institute, University of Queensland.,Australian e-Health Research Centre, CSIRO, Herston, Australia.,Department of Psychology, James Cook University
| | - Dajiang Zhu
- Department of Computer Science and Engineering, The University of Texas at Arlington
| | | | | | | | | | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Greig I de Zubicaray
- Institute of Health and Biomedical Innovation, Queensland University of Technology
| | - Katie L McMahon
- Herston Imaging Research Facility and School of Clinical Sciences, Queensland University of Technology
| | - Margaret J Wright
- Queensland Brain Institute, University of Queensland.,Centre for Advanced Imaging, University of Queensland
| | - Brendan P Zietsch
- Centre for Psychology and Evolution, School of Psychology, University of Queensland
| |
Collapse
|
82
|
Haase J, Jones AKC, Mc Veigh CJ, Brown E, Clarke G, Ahnert-Hilger G. Sex and brain region-specific regulation of serotonin transporter activity in synaptosomes in guanine nucleotide-binding protein G(q) alpha knockout mice. J Neurochem 2021; 159:156-171. [PMID: 34309872 DOI: 10.1111/jnc.15482] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
The regulation of the serotonin transporter (SERT) by guanine nucleotide-binding protein alpha (Gα) q was investigated using Gαq knockout mice. In the absence of Gαq, SERT-mediated uptake of 5-hydroxytryptamine (5HT) was enhanced in midbrain and frontal cortex synaptosomes, but only in female mice. The mechanisms underlying this sexual dimorphism were investigated using quantitative western blot analysis revealing brain region-specific differences. In the frontal cortex, SERT protein expression was decreased in male knockout mice, seemingly explaining the sex-dependent variation in SERT activity. The differential expression of Gαi1 in female mice contributes to the sex differences in the midbrain. In fact, Gαi1 levels inversely correlate with 5HT uptake rates across both sexes and genotypes. Likely due to differential SERT regulation as well as sex differences in the expression of tryptophan hydroxylase 2, Gαq knockout mice also displayed sex- and genotype-dependent alterations in total 5HT tissue levels as determined by high-performance liquid chromatography. Gαq inhibitors, YM-254890 and BIM-46187, differentially affected SERT activity in both, synaptosomes and cultured cells. YM-254890 treatment mimicked the effect of Gαq knockout in the frontal cortex. BIM-46187, which promotes the nucleotide-free form of Gα proteins, substantially inhibited 5HT uptake, prompting us to hypothesise that Gαq interacts with SERT similarly as with G-protein-coupled receptors and inhibits SERT activity by modulating transport-associated conformational changes. Taken together, our findings reveal a novel mechanism of SERT regulation and impact our understanding of sex differences in diseases associated with dysregulation of serotonin transmission, such as depression and anxiety.
Collapse
Affiliation(s)
- Jana Haase
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Aimée K C Jones
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Conor J Mc Veigh
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Eric Brown
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland and Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Gudrun Ahnert-Hilger
- Institute of Integrative Neuroanatomy, Charité University Medicine Berlin and Max-Planck-Institute for Biophysical Chemistry Göttingen, Göttingen, Germany
| |
Collapse
|
83
|
Hecht EE, Kukekova AV, Gutman DA, Acland GM, Preuss TM, Trut LN. Neuromorphological Changes following Selection for Tameness and Aggression in the Russian Farm-Fox experiment. J Neurosci 2021; 41:6144-6156. [PMID: 34127519 PMCID: PMC8276742 DOI: 10.1523/jneurosci.3114-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 01/13/2023] Open
Abstract
The Russian farm-fox experiment is an unusually long-running and well-controlled study designed to replicate wolf-to-dog domestication. As such, it offers an unprecedented window onto the neural mechanisms governing the evolution of behavior. Here we report evolved changes to gray matter morphology resulting from selection for tameness versus aggressive responses toward humans in a sample of 30 male fox brains. Contrasting with standing ideas on the effects of domestication on brain size, tame foxes did not show reduced brain volume. Rather, gray matter volume in both the tame and aggressive strains was increased relative to conventional farm foxes bred without deliberate selection on behavior. Furthermore, tame- and aggressive-enlarged regions overlapped substantially, including portions of motor, somatosensory, and prefrontal cortex, amygdala, hippocampus, and cerebellum. We also observed differential morphologic covariation across distributed gray matter networks. In one prefrontal-cerebellum network, this covariation differentiated the three populations along the tame-aggressive behavioral axis. Surprisingly, a prefrontal-hypothalamic network differentiated the tame and aggressive foxes together from the conventional strain. These findings indicate that selection for opposite behaviors can influence brain morphology in a similar way.SIGNIFICANCE STATEMENT Domestication represents one of the largest and most rapid evolutionary shifts of life on earth. However, its neural correlates are largely unknown. Here we report the neuroanatomical consequences of selective breeding for tameness or aggression in the seminal Russian farm-fox experiment. Compared with a population of conventional farm-bred control foxes, tame foxes show neuroanatomical changes in the PFC and hypothalamus, paralleling wolf-to-dog shifts. Surprisingly, though, aggressive foxes also show similar changes. Moreover, both strains show increased gray matter volume relative to controls. These results indicate that similar brain adaptations can result from selection for opposite behavior, that existing ideas of brain changes in domestication may need revision, and that significant neuroanatomical change can evolve very quickly, within the span of <100 generations.
Collapse
Affiliation(s)
- Erin E Hecht
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA 02138
| | - Anna V Kukekova
- Department of Animal Sciences, College of Agriculture, Consumer, and Environmental Sciences, University of IL Urbana-Champaign, Urbana, IL 61801
| | | | - Gregory M Acland
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, 14853
| | - Todd M Preuss
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30322
| | - Lyudmila N Trut
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia, 630090
| |
Collapse
|
84
|
Turano A, McAuley EM, Muench MC, Schwarz JM. EXAMINING THE IMPACT OF NEUROIMMUNE DYSREGULATION ON SOCIAL BEHAVIOR OF MALE AND FEMALE JUVENILE RATS. Behav Brain Res 2021; 415:113449. [PMID: 34252501 DOI: 10.1016/j.bbr.2021.113449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 11/29/2022]
Abstract
Many individuals diagnosed with neuropsychiatric disorders, such as autism, attention-deficit/hyperactivity disorder, schizophrenia, and social anxiety disorder, all share a common dimension of aberrant social behavior. Epidemiological data indicate that adverse environmental factors contribute to the risk for neurodevelopmental disorders, including those associated with aberrant social behavior. Early-life exposure to infectious pathogens is one of those adverse environmental factors, suggesting that activation of the immune system during early development may contribute to disease pathology associated with altered social behavior. In the current project, we examined the impact of neonatal infection, with or without juvenile immune activation, on the expression of juvenile social behavior and on the expression of inflammatory cytokines and microglial signaling molecules in the juvenile rat brain. The outcomes of these experiments revealed that neonatal infection significantly decreased juvenile social interaction, but significantly increased juvenile play behavior in male and female rats. Moreover, neonatal infection alone, juvenile immune activation alone, and neonatal infection plus juvenile immune activation all significantly impaired social recognition in juvenile male rats. Juvenile female rats (including controls) failed to demonstrate social recognition as measured in our three-chamber social recognition test. Taken together, the behavioral and molecular data presented here support the sensitivity of the developing brain to immune activation, particularly in the expression of age-appropriate social behaviors. These data warrant the design of additional studies to examine the mechanistic relationship between early-life immune activation and aberrant social behavior to develop novel as well as modify existing therapeutic targets and preventative measures to help those who display aberrant social behavior.
Collapse
Affiliation(s)
- Alexandra Turano
- University of Delaware, Department of Psychological and Brain Sciences, 105 The Green, Newark, Delaware, 19716, United States.
| | - Elizabeth M McAuley
- University of Delaware, Department of Psychological and Brain Sciences, 105 The Green, Newark, Delaware, 19716, United States.
| | - Megan C Muench
- University of Delaware, Department of Psychological and Brain Sciences, 105 The Green, Newark, Delaware, 19716, United States.
| | - Jaclyn M Schwarz
- University of Delaware, Department of Psychological and Brain Sciences, 105 The Green, Newark, Delaware, 19716, United States.
| |
Collapse
|
85
|
Brennan D, Wu T, Fan J. Morphometrical Brain Markers of Sex Difference. Cereb Cortex 2021; 31:3641-3649. [PMID: 33774662 DOI: 10.1093/cercor/bhab037] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 01/22/2021] [Accepted: 02/06/2021] [Indexed: 01/02/2023] Open
Abstract
Many major neuropsychiatric pathologies, some of which appear in adolescence, show differentiated prevalence, onset, and symptomatology across the biological sexes. Therefore, mapping differences in brain structure between males and females during this critical developmental period may provide information about the neural mechanisms underlying the dimorphism of these pathologies. Utilizing a large dataset collected through the Adolescent Brain Cognitive Development study, we investigated the differences of adolescent (9-10 years old) male and female brains (n = 8325) by using a linear Support-Vector Machine Classifier to predict sex based on morphometry and image intensity values of structural brain imaging data. The classifier correctly classified the sex of 86% individuals with the insula, the precentral and postcentral gyri, and the pericallosal sulcus as the most discernable features. These results demonstrate the existence of complex, yet robustly measurable morphometrical brain markers of sex difference.
Collapse
Affiliation(s)
- Daniel Brennan
- Department of Psychology, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Tingting Wu
- Department of Psychology, Queens College, The City University of New York, Queens, NY 11367, USA
| | - Jin Fan
- Department of Psychology, Queens College, The City University of New York, Queens, NY 11367, USA
| |
Collapse
|
86
|
Abstract
In contrast to traditional laboratory animals, prairie voles form socially monogamous partnerships in the wild and exhibit lasting social preferences for familiar individuals-both mates and same-sex peers-in the laboratory. Decades of research into the mechanisms supporting pair bonding behavior have made prairie voles an important model organism for the study of social relationships. The partner preference test is a laboratory test of familiarity preference that takes place over an extended interval (typically 3 hr), during which test subjects can directly interact with conspecifics and often engage in resting side-by-side contact (i.e., huddling). The use of this test has enabled study of the neural pathways and mechanisms involved in promoting or impairing relationship formation. The tendency to form partner preferences is also used as a behavioral indicator of the effects of early life experiences and environmental exposures. While this test was developed to assess the extent of social preference for mates in prairie voles, it has been adapted for use in other social contexts and in multiple other species. This article provides instructions for conducting the classic partner preference test, as well as variations including same-sex "peer" partner preference tests. The effects of several protocol variations are examined, including duration of cohousing, separation interval, use of tethers versus barriers, linear versus branched apparatus configuration, and duration of the test. The roles of social variables including sex of the focal individual, sex of conspecifics, reproductive state, and use of the test in other species are then considered. Finally, sample data are provided along with discussion of scoring and statistical analysis of partner preference tests. © 2021 Wiley Periodicals LLC. Basic Protocol: Partner preference test Support Protocol: Behavioral scoring.
Collapse
|
87
|
Abstract
Prairie voles have emerged as an important rodent model for understanding the neuroscience of social behavior. Prairie voles are well known for their capacity for pair bonding and alloparental care. These behavioral phenomena overlap with human social behavior but are not commonly observed in traditional rodent models. In this article, we highlight the many benefits of using prairie voles in neuroscience research. We begin by describing the advantages of using diverse and non-traditional study models. We then focus on social behaviors, including pair bonding, alloparental care, and peer interactions, that have brought voles to the forefront of social neuroscience. We describe many additional features of prairie vole biology and behavior that provide researchers with opportunities to address an array of research questions. We also survey neuroethological methods that have been used with prairie voles, from classic to modern techniques. Finally, we conclude with a discussion of other vole species, particularly meadow voles, and their own unique advantages for neuroscience studies. This article provides a foundation for researchers who are new to working with voles, as well as for experienced neuroscientists who want to expand their research scope. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- William M. Kenkel
- Department of Psychological & Brain Sciences, University of Delaware, Newark, DE 19716
| | - Morgan L. Gustison
- Department of Integrative Biology, University of Texas at Austin, Austin, TX 78712 USA
| | - Annaliese K. Beery
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720 USA
| |
Collapse
|
88
|
Bhargava A, Arnold AP, Bangasser DA, Denton KM, Gupta A, Hilliard Krause LM, Mayer EA, McCarthy M, Miller WL, Raznahan A, Verma R. Considering Sex as a Biological Variable in Basic and Clinical Studies: An Endocrine Society Scientific Statement. Endocr Rev 2021; 42:219-258. [PMID: 33704446 PMCID: PMC8348944 DOI: 10.1210/endrev/bnaa034] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Indexed: 02/08/2023]
Abstract
In May 2014, the National Institutes of Health (NIH) stated its intent to "require applicants to consider sex as a biological variable (SABV) in the design and analysis of NIH-funded research involving animals and cells." Since then, proposed research plans that include animals routinely state that both sexes/genders will be used; however, in many instances, researchers and reviewers are at a loss about the issue of sex differences. Moreover, the terms sex and gender are used interchangeably by many researchers, further complicating the issue. In addition, the sex or gender of the researcher might influence study outcomes, especially those concerning behavioral studies, in both animals and humans. The act of observation may change the outcome (the "observer effect") and any experimental manipulation, no matter how well-controlled, is subject to it. This is nowhere more applicable than in physiology and behavior. The sex of established cultured cell lines is another issue, in addition to aneuploidy; chromosomal numbers can change as cells are passaged. Additionally, culture medium contains steroids, growth hormone, and insulin that might influence expression of various genes. These issues often are not taken into account, determined, or even considered. Issues pertaining to the "sex" of cultured cells are beyond the scope of this Statement. However, we will discuss the factors that influence sex and gender in both basic research (that using animal models) and clinical research (that involving human subjects), as well as in some areas of science where sex differences are routinely studied. Sex differences in baseline physiology and associated mechanisms form the foundation for understanding sex differences in diseases pathology, treatments, and outcomes. The purpose of this Statement is to highlight lessons learned, caveats, and what to consider when evaluating data pertaining to sex differences, using 3 areas of research as examples; it is not intended to serve as a guideline for research design.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Obstetrics and Gynecology, University of California, San Francisco, CA, USA
| | - Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Arpana Gupta
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Emeran A Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Division of Digestive Diseases, University of California, Los Angeles, Los Angeles, CA, USA
| | - Margaret McCarthy
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Walter L Miller
- Center for Reproductive Sciences, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institutes of Mental Health, Intramural Research Program, Bethesda, MD, USA
| | - Ragini Verma
- Diffusion and Connectomics In Precision Healthcare Research (DiCIPHR) lab, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
89
|
Joye DAM, Evans JA. Sex differences in daily timekeeping and circadian clock circuits. Semin Cell Dev Biol 2021; 126:45-55. [PMID: 33994299 DOI: 10.1016/j.semcdb.2021.04.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/24/2021] [Accepted: 04/29/2021] [Indexed: 11/19/2022]
Abstract
The circadian system regulates behavior and physiology in many ways important for health. Circadian rhythms are expressed by nearly every cell in the body, and this large system is coordinated by a central clock in the suprachiasmatic nucleus (SCN). Sex differences in daily rhythms are evident in humans and understanding how circadian function is modulated by biological sex is an important goal. This review highlights work examining effects of sex and gonadal hormones on daily rhythms, with a focus on behavior and SCN circuitry in animal models commonly used in pre-clinical studies. Many questions remain in this area of the field, which would benefit from further work investigating this topic.
Collapse
Affiliation(s)
- Deborah A M Joye
- Marquette University, Department of Biomedical Sciences, Milwaukee, WI, USA
| | - Jennifer A Evans
- Marquette University, Department of Biomedical Sciences, Milwaukee, WI, USA.
| |
Collapse
|
90
|
Impact of intrauterine fetal resuscitation with oxygen on oxidative stress in the developing rat brain. Sci Rep 2021; 11:9798. [PMID: 33963277 PMCID: PMC8105387 DOI: 10.1038/s41598-021-89299-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022] Open
Abstract
Use of maternal oxygen for intrauterine resuscitation is contentious because of the lack of evidence for its efficacy and the possibility of fetal harm through oxidative stress. Because the developing brain is rich in lipids and low in antioxidants, it remains vulnerable to oxidative stress. Here, we tested this hypothesis in a term pregnant rat model with oxytocin-induced fetal distress followed by treatment with either room air or 100% oxygen for 6 h. Fetal brains from both sexes were subjected to assays for biomarkers of oxidative stress (4-hydroxynonenal, protein carbonyl, or 8-hydroxy-2'-deoxyguanosine), expression of genes mediating oxidative stress, and mitochondrial oxidative phosphorylation. Contrary to our hypothesis, maternal hyperoxia was not associated with increased biomarkers of oxidative stress in the fetal brain. However, there was significant upregulation of the expression of select genes mediating oxidative stress, of which some were male-specific. These observations, however, were not accompanied by changes in the expression of proteins from the mitochondrial electron transport chain. In summary, maternal hyperoxia in the setting of acute uteroplacental ischemia-hypoxia does not appear to cause oxidative damage to the developing brain.
Collapse
|
91
|
Wahlstrom KL, Alvarez-Dieppa AC, McIntyre CK, LaLumiere RT. The medial entorhinal cortex mediates basolateral amygdala effects on spatial memory and downstream activity-regulated cytoskeletal-associated protein expression. Neuropsychopharmacology 2021; 46:1172-1182. [PMID: 33007779 PMCID: PMC8115646 DOI: 10.1038/s41386-020-00875-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/08/2020] [Accepted: 09/21/2020] [Indexed: 01/09/2023]
Abstract
The basolateral amygdala (BLA) modulates the consolidation of dorsal hippocampus (DH)-dependent spatial and dorsolateral striatum (DLS)-dependent cued-response memories, often in competition with one another. Evidence suggests that a critical mechanism for BLA influences on memory consolidation is via effects on activity-regulated cytoskeletal-associated protein (ARC) in downstream brain regions. However, the circuitry by which the BLA modulates ARC in multiple competing memory systems remains unclear. Prior evidence indicates that optogenetic stimulation of BLA projections to the medial entorhinal cortex (mEC) enhances the consolidation of spatial learning and impairs the consolidation of cued-response learning, suggesting this pathway provides a circuit for favoring one system over another. Therefore, we hypothesized the BLA-mEC pathway mediates effects on downstream ARC-based synaptic plasticity related to these competing memory systems. To address this, male and female Sprague-Dawley rats underwent spatial or cued-response Barnes maze training and, 45 min later, were sacrificed for ARC analysis in synaptoneurosomes from the DH and DLS. Initial experiments found that spatial training alone increased ARC levels in the DH above those observed in control rats and rats that underwent a cued-response version of the task. Postspatial training optogenetic stimulation of the BLA-mEC pathway altered the balance of ARC expression in the DH vs. DLS, specifically shifting the balance in favor of the DH-based spatial memory system, although the precise region of ARC changes differed by sex. These findings suggest that BLA-mEC pathway influences on ARC in downstream regions are a mechanism by which the BLA can favor one memory system over another.
Collapse
Affiliation(s)
- Krista L. Wahlstrom
- grid.214572.70000 0004 1936 8294Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242 USA
| | - Amanda C. Alvarez-Dieppa
- grid.267323.10000 0001 2151 7939School of Behavioral and Brain Sciences, University of Texas-Dallas, Richardson, TX 75080 USA
| | - Christa K. McIntyre
- grid.267323.10000 0001 2151 7939School of Behavioral and Brain Sciences, University of Texas-Dallas, Richardson, TX 75080 USA
| | - Ryan T. LaLumiere
- grid.214572.70000 0004 1936 8294Department of Psychological and Brain Sciences, University of Iowa, Iowa City, IA 52242 USA ,grid.214572.70000 0004 1936 8294Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242 USA
| |
Collapse
|
92
|
Shansky RM, Murphy AZ. Considering sex as a biological variable will require a global shift in science culture. Nat Neurosci 2021; 24:457-464. [PMID: 33649507 DOI: 10.1038/s41593-021-00806-8] [Citation(s) in RCA: 223] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/19/2021] [Indexed: 01/31/2023]
Abstract
For over half a century, male rodents have been the default model organism in preclinical neuroscience research, a convention that has likely contributed to higher rates of misdiagnosis and adverse side effects from drug treatment in women. Studying both sexes could help to rectify these public health problems, but incentive structures in publishing and career advancement deter many researchers from doing so. Moreover, funding agency directives to include male and female animals and human participants in grant proposals lack mechanisms to hold recipients accountable. In this Perspective, we highlight areas of behavioral, cellular and systems neuroscience in which fundamental sex differences have been identified, demonstrating that truly rigorous science must include males and females. We call for a cultural and structural change in how we conduct research and evaluate scientific progress, realigning our professional reward systems and experimental standards to produce a more equitable, representative and therefore translational body of knowledge.
Collapse
Affiliation(s)
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
93
|
Lolier M, Wagner CK. Sex differences in dopamine innervation and microglia are altered by synthetic progestin in neonatal medial prefrontal cortex. J Neuroendocrinol 2021; 33:e12962. [PMID: 33719165 PMCID: PMC8130850 DOI: 10.1111/jne.12962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/06/2021] [Accepted: 02/18/2021] [Indexed: 12/16/2022]
Abstract
The synthetic progestin 17-α-hydroxyprogesterone caproate (17-OHPC) is commonly prescribed to pregnant women with a history of preterm delivery, despite little evidence of efficacy. The timing of 17-OHPC administration coincides with fetal mesocortical dopamine pathway development, yet the potential effects on cortical development and cognition are almost unknown. In rodent models, exposure to 17-OHPC significantly increased dopaminergic innervation of the medial prefrontal cortex (mPFC), an aberrant pattern of connectivity that may underlie deficits in cognitive flexibility observed in adulthood. In the present study, tyrosine hydroxylase (TH) immunoreactivity was used to determine whether 17-OHPC altered dopaminergic innervation of the mPFC during a neonatal period of synaptogenesis in males and females. Although there were no differences in the amount of TH-immunoreactive (-IR) fibres, there was a sex difference in TH-IR fibre distribution in deep layers of the prelimbic area (PL) mPFC; males had a narrower pattern of dopaminergic innervation than females. 17-OHPC exposure abolished these sex-specific patterns, such that 17-OHPC females had a narrower pattern in the PL than control females. In the infralimbic mPFC (IL), 17-OHPC males had a broader pattern of distribution of TH-immunoreactivity than control males with no differences in the amount of TH-IR fibres. 17-OHPC also created a sex difference in which males had a lower TH-IR fibre density than females. We also examined microglia, brain macrophages that play a key role in sculpting dopaminergic axon outgrowth in development, using phenotype as an indirect measure of microglial activity. Females had a greater number of reactive stout microglia compared to males in the PL, and males had more active round microglia than females in the IL. 17-OHPC treatment abolished the sex differences in both regions. These findings demonstrate that developmental exposure to 17-OHPC can exert differential effects in males and females and may diminish sex differences in cortical maturation.
Collapse
Affiliation(s)
- Melanie Lolier
- Department of Psychology & Center for Neuroscience Research, University at Albany, Albany, NY, USA
| | - Christine K Wagner
- Department of Psychology & Center for Neuroscience Research, University at Albany, Albany, NY, USA
| |
Collapse
|
94
|
Dump the "dimorphism": Comprehensive synthesis of human brain studies reveals few male-female differences beyond size. Neurosci Biobehav Rev 2021; 125:667-697. [PMID: 33621637 DOI: 10.1016/j.neubiorev.2021.02.026] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 01/01/2021] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
With the explosion of neuroimaging, differences between male and female brains have been exhaustively analyzed. Here we synthesize three decades of human MRI and postmortem data, emphasizing meta-analyses and other large studies, which collectively reveal few reliable sex/gender differences and a history of unreplicated claims. Males' brains are larger than females' from birth, stabilizing around 11 % in adults. This size difference accounts for other reproducible findings: higher white/gray matter ratio, intra- versus interhemispheric connectivity, and regional cortical and subcortical volumes in males. But when structural and lateralization differences are present independent of size, sex/gender explains only about 1% of total variance. Connectome differences and multivariate sex/gender prediction are largely based on brain size, and perform poorly across diverse populations. Task-based fMRI has especially failed to find reproducible activation differences between men and women in verbal, spatial or emotion processing due to high rates of false discovery. Overall, male/female brain differences appear trivial and population-specific. The human brain is not "sexually dimorphic."
Collapse
|
95
|
Cahill L. It's time to move past biases against sex differences research: Commentary on Spets and Slotnick. Cogn Neurosci 2021; 12:174-175. [PMID: 33416033 DOI: 10.1080/17588928.2020.1867085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Neuroscience is uncovering sex influences at all levels of mammalian brain function at an accelerating rate. Unfortunately, persistent biases against the topic remain among some investigators. One is that sex influences are small and unreliable, despite the existence of no evidence supporting this general assertion. In this volume, Spets and Slotnick provide clear evidence for a consistent sex influence on one aspect of human cognition, retrieval from long-term memory.
Collapse
Affiliation(s)
- Larry Cahill
- Department of Neurobiology and Behavior, University of California, McGaugh Hall, Irvine
| |
Collapse
|
96
|
Liang X, Cheng S, Ye J, Chu X, Wen Y, Liu L, Qi X, Jia Y, Zhang F. Evaluating the genetic effects of sex hormone traits on the development of mental traits: a polygenic score analysis and gene-environment-wide interaction study in UK Biobank cohort. Mol Brain 2021; 14:3. [PMID: 33407712 PMCID: PMC7788797 DOI: 10.1186/s13041-020-00718-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 12/15/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To evaluate the genetic effects of sex hormone traits on the development of mental traits in middle-aged adults. METHODS The SNPs associated with sex hormone traits were derived from a two-stage genome-wide association study (GWAS). Four sex hormone traits were selected in the current study, including sex hormone-binding globulin (SHBG), testosterone, bioavailable testosterone and estradiol. The polygenic risk score (PRS) of sex hormone traits were calculated from individual-level genotype data of the United Kingdom (UK) Biobank cohort. We then used logistic and linear regression models to assess the associations between individual PRS of sex hormone traits and the frequency of alcohol consumption, anxiety, intelligence and so on. Finally, gene-environment-wide interaction study (GEWIS) was performed to detect novel candidate genes interacting with the sex hormone traits on the development of fluid intelligence and the frequency of smoking and alcohol consumption by PLINK2.0. RESULTS We observed positive association between SHBG and the frequency of alcohol consumption (b = 0.0101, p = 3.84 × 10-11) in middle-aged males and females. In addition, estradiol was positively associated with the frequency of alcohol consumption (b = 0.0128, p = 1.96 × 10-8) in middle-aged males. Moreover, bioavailable testosterone was associated with the fluid intelligence (b = - 0.0136, p = 5.74 × 10-5) in middle-aged females. Finally, GEWIS identified one significant loci, Tenascin R (TNR) (rs34633780, p = 3.45 × 10-8) interacting with total testosterone for fluid intelligence. CONCLUSION Our study results support the genetic effects of sex hormone traits on the development of intelligence and the frequency of alcohol consumption in middle-aged adults in UK.
Collapse
Affiliation(s)
- Xiao Liang
- National Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - ShiQiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - Jing Ye
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - XiaoMeng Chu
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - Xin Qi
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - YuMeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, 71006, China.
| |
Collapse
|
97
|
Loss of α7 nicotinic acetylcholine receptors in GABAergic neurons causes sex-dependent decreases in radial glia-like cell quantity and impairments in cognitive and social behavior. Brain Struct Funct 2021; 226:365-379. [PMID: 33398432 DOI: 10.1007/s00429-020-02179-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
The dentate gyrus (DG) is a unique brain structure in that neurons can be generated postnatally and integrated within existing circuitry throughout life. The maturation process of these newly generated neurons (granule cells) is modulated by nicotinic acetylcholine receptors (nAChRs) through a variety of mechanisms such as neural stem pool proliferation, cell survival, signal modulation, and dendritic integration. Disrupted nAChR signaling has been implicated in neuropsychiatric and neurodegenerative disorders, potentially via alterations in DG neurogenesis. GABAergic interneurons are known to express nAChRs, predominantly the α7 subtype, and have been shown to shape development, integration, and circuit reorganization of DG granule cells. Therefore, we examined histological and behavioral effects of knocking out α7 nAChRs in GABAergic neurons. Deletion of α7 nAChRs resulted in a reduction of radial glia-like cells within the subgranular zone of the DG and a concomitant trend towards decreased immature neurons, specifically in male mice, as well as sex-dependent changes in several behaviors, including social recognition and spatial learning. Overall, these findings suggest α7 nAChRs expressed in GABAergic neurons play an important role in regulating the adult neural stem cell pool and behavior in a sex-dependent manner. This provides important insight into the mechanisms by which cholinergic dysfunction contributes to the cognitive and behavioral changes associated with neurodevelopmental and neurodegenerative disorders.
Collapse
|
98
|
Rurak GM, Woodside B, Aguilar-Valles A, Salmaso N. Astroglial cells as neuroendocrine targets in forebrain development: Implications for sex differences in psychiatric disease. Front Neuroendocrinol 2021; 60:100897. [PMID: 33359797 DOI: 10.1016/j.yfrne.2020.100897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most abundant cell type in the mammalian brain. They are implicated in almost every aspect of brain physiology, including maintaining homeostasis, building and maintaining the blood brain barrier, and the development and maturation of neuronal networks. Critically, astroglia also express receptors for gonadal sex hormones, respond rapidly to gonadal hormones, and are able to synthesize hormones. Thus, they are positioned to guide and mediate sexual differentiation of the brain, particularly neuronal networks in typical and pathological conditions. In this review, we describe astroglial involvement in the organization and development of the brain, and consider known sex differences in astroglial responses to understand how astroglial cell-mediated organization may play a role in forebrain sexual dimorphisms in human populations. Finally, we consider how sexually dimorphic astroglial responses and functions in development may lead to sex differences in vulnerability for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; Concordia University, Montreal, Quebec, Canada
| | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
99
|
Reppucci CJ, Brown LA, Chambers AQ, Veenema AH. Wistar rats and C57BL/6 mice differ in their motivation to seek social interaction versus food in the Social versus Food Preference Test. Physiol Behav 2020; 227:113162. [PMID: 32877644 PMCID: PMC7655716 DOI: 10.1016/j.physbeh.2020.113162] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/08/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023]
Abstract
Here we characterized the Social versus Food Preference Test, a behavioral paradigm designed to investigate the competition between the choice to seek social interaction versus the choice to seek food. We assessed how this competition was modulated by internal cues (social isolation, food deprivation), external cues (stimulus salience), sex (males, females), age (adolescents, adults), and rodent model (Wistar rats, C57BL/6 mice). We found that changes in stimulus preference in response to the internal and external cue manipulations were similar across cohorts. Specifically, social over food preference scores were reduced by food deprivation and social familiarly in Wistar rats and C57BL/6 mice of both sexes. Interestingly, the degree of food deprivation-induced changes in stimulus investigation patterns were greater in adolescents compared to adults in Wistar rats and C57BL/6 mice. Strikingly, baseline stimulus preference and investigation times varied greatly between rodent models: across manipulations, Wistar rats were generally more social-preferring and C57BL/6 mice were generally more food-preferring. Adolescent Wistar rats spent more time investigating the social and food stimuli than adult Wistar rats, while adolescent and adult C57BL/6 mice investigated the stimuli a similar amount. Social isolation did not alter behavior in the Social versus Food Preference Test. Together, our results indicate that the Social versus Food Preference Test is a flexible behavioral paradigm suitable for future interrogations of the peripheral and central systems that can coordinate the expression of stimulus preference related to multiple motivated behaviors.
Collapse
Affiliation(s)
- Christina J Reppucci
- Department of Psychology & Neuroscience Program, Michigan State University, 766 Service Road, 4016 ISTB, East Lansing, MI 48824, United States.
| | - Leigha A Brown
- Department of Psychology & Neuroscience Program, Michigan State University, 766 Service Road, 4016 ISTB, East Lansing, MI 48824, United States
| | - Ashley Q Chambers
- Department of Psychology & Neuroscience Program, Michigan State University, 766 Service Road, 4016 ISTB, East Lansing, MI 48824, United States
| | - Alexa H Veenema
- Department of Psychology & Neuroscience Program, Michigan State University, 766 Service Road, 4016 ISTB, East Lansing, MI 48824, United States
| |
Collapse
|
100
|
Arnold AP. Four Core Genotypes and XY* mouse models: Update on impact on SABV research. Neurosci Biobehav Rev 2020; 119:1-8. [PMID: 32980399 PMCID: PMC7736196 DOI: 10.1016/j.neubiorev.2020.09.021] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 09/13/2020] [Accepted: 09/15/2020] [Indexed: 12/17/2022]
Abstract
The impact of two mouse models is reviewed, the Four Core Genotypes and XY* models. The models are useful for determining if the causes of sex differences in phenotypes are either hormonal or sex chromosomal, or both. Used together, the models also can distinguish between the effects of X or Y chromosome genes that contribute to sex differences in phenotypes. To date, the models have been used to uncover sex chromosome contributions to sex differences in a wide variety of phenotypes, including brain and behavior, autoimmunity and immunity, cardiovascular disease, metabolism, and Alzheimer's Disease. In some cases, use of the models has been a strategy leading to discovery of specific X or Y genes that protect from or exacerbate disease. Sex chromosome and hormonal factors interact, in some cases to reduce the effects of each other. Future progress will come from more extensive application of these models, and development of similar models in other species.
Collapse
Affiliation(s)
- Arthur P Arnold
- Department of Integrative Biology & Physiology, Laboratory of Neuroendocrinology of the Brain Research Institute, University of California, UCLA, 610 Charles Young Drive South, Los Angeles, CA, 90095-7239, United States.
| |
Collapse
|