1
|
Hatkevich T, Wilhelm D. Editorial: Proceedings of the 9th international symposium on the biology of vertebrate sex determination 2023. Front Cell Dev Biol 2024; 12:1530367. [PMID: 39703695 PMCID: PMC11655486 DOI: 10.3389/fcell.2024.1530367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024] Open
Affiliation(s)
- Talia Hatkevich
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - Dagmar Wilhelm
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Robinson CD, Hale MD, Cox CL, John-Alder HB, Cox RM. Effects of Testosterone on Gene Expression Are Concordant between Sexes but Divergent across Species of Sceloporus Lizards. Am Nat 2024; 204:517-532. [PMID: 39486031 DOI: 10.1086/732200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
AbstractHormones mediate sexual dimorphism by regulating sex-specific patterns of gene expression, but it is unclear how much of this regulation involves sex-specific hormone levels versus sex-specific transcriptomic responses to the same hormonal signal. Moreover, transcriptomic responses to hormones can evolve, but the extent to which hormonal pleiotropy in gene regulation is conserved across closely related species is not well understood. We addressed these issues by elevating testosterone levels in juvenile females and males of three Sceloporus lizard species before sexual divergence in circulating testosterone and then characterizing transcriptomic responses in the liver. In each species, more genes were responsive to testosterone in males than in females, suggesting that early developmental processes prime sex-specific transcriptomic responses to testosterone later in life. However, overall transcriptomic responses to testosterone were concordant between sexes, with no genes exhibiting sex-by-treatment interactions. By contrast, hundreds of genes exhibited species-by-treatment interactions, particularly when comparing distantly related species with different patterns of sexual dimorphism, suggesting evolutionary lability in gene regulation by testosterone. Collectively, our results indicate that early organizational effects may lead to sex-specific differences in the magnitude, but not the direction, of transcriptomic responses to testosterone and that the hormone-genome interface accrues regulatory changes over evolutionary time.
Collapse
|
3
|
Jang GF, Crabb JS, Grenell A, Wolk A, Campla C, Luo S, Ali M, Hu B, Willard B, Anand-Apte B. Quantitative proteomic profiling reveals sexual dimorphism in the retina and RPE of C57BL6 mice. Biol Sex Differ 2024; 15:87. [PMID: 39478535 PMCID: PMC11526624 DOI: 10.1186/s13293-024-00645-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 08/21/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Sex as a biological variable is not a common consideration in molecular mechanistic or preclinical studies of retinal diseases. Understanding the sexual dimorphism of adult RPE and retina under physiological conditions is an important first step in improving our understanding of sex-based physio-pathological mechanisms. METHODS Isobaric tags for relative and absolute quantitation (iTRAQ) were used for quantitative proteomics of male and female mouse retina and RPE (10 mice of each sex for each tissue type). Differentially expressed proteins were subjected to Gene Ontology (GO) analysis and Ingenuity Pathway Analysis (IPA). RESULTS Differential expression analysis identified 21 differentially expressed proteins in the retina and 58 differentially expressed proteins in the RPE. Ingenuity pathway analysis identified the top canonical pathways differentially activated in the retina to be calcium transport I, nucleotide excision repair, molecular transport and cell death and survival. In the RPE, the top canonical pathways were calcium signaling, dilated cardiomyopathy signaling, actin cytoskeletal signaling and cellular assembly and organization. CONCLUSIONS These results provide insights into sex differences in the retina and RPE proteome of mice and begin to shed clues into the sexual dimorphism seen in retinal diseases.
Collapse
Affiliation(s)
- Geeng-Fu Jang
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John S Crabb
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Allison Grenell
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Alyson Wolk
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christie Campla
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Shiming Luo
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mariya Ali
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Belinda Willard
- Mass Spectrometry Laboratory for Protein Sequencing, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bela Anand-Apte
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
4
|
Malcore RM, Samanta MK, Kalantry S, Iwase S. Regulation of Sex-biased Gene Expression by the Ancestral X-Y Chromosomal Gene Pair Kdm5c-Kdm5d. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620066. [PMID: 39484414 PMCID: PMC11527134 DOI: 10.1101/2024.10.24.620066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Conventionally, Y-linked Sry is thought to drive sex differences by triggering differential hormone production. Ancestral X-Y gene pairs, however, are hypothesized to drive hormone-independent sex differences. Here, we show that the X-Y gene pair Kdm5c-Kdm5d regulates sex-biased gene expression in pluripotent mouse embryonic stem cells (ESCs). Wild-type (WT) XX female ESCs exhibit >2-fold higher expression of 409 genes relative to WT XY male ESCs. Conversely, WT XY male ESCs exhibit >2-fold higher expression of 126 genes compared to WT XX female ESCs. Loss of Kdm5c in female ESCs downregulates female-biased genes. In contrast, loss of either Kdm5c or Kdm5d in male ESCs upregulates female-biased genes and downregulates male-biased genes, effectively neutralizing sex-biased gene expression. KDM5C promotes the expression of Kdm5d and several other Y-linked genes in male ESCs. Moreover, ectopic Kdm5d expression in female ESCs is sufficient to drive male-biased gene expression. These results establish Kdm5c-Kdm5d as critical regulators of sex-biased gene expression.
Collapse
Affiliation(s)
- Rebecca M. Malcore
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Milan Kumar Samanta
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| | - Sundeep Kalantry
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105, USA
- Lead contact
| | - Shigeki Iwase
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48105, USA
| |
Collapse
|
5
|
Torgerson C, Bottenhorn K, Ahmadi H, Choupan J, Herting MM. More similarity than difference: comparison of within- and between-sex variance in early adolescent brain structure. RESEARCH SQUARE 2024:rs.3.rs-4947186. [PMID: 39483919 PMCID: PMC11527358 DOI: 10.21203/rs.3.rs-4947186/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background Adolescent neuroimaging studies of sex differences in the human brain predominantly examine mean differences between males and females. This focus on between-groups differences without probing relative distributions and similarities may contribute to both conflation and overestimation of sex differences and sexual dimorphism in the developing human brain. Methods We aimed to characterize the variance in brain macro- and micro-structure in early adolescence as it pertains to sex at birth using a large sample of 9-11 year-olds from the Adolescent Brain Cognitive Development (ABCD) Study (N=7,723). Specifically, for global and regional estimates of gray and white matter volume, cortical thickness, and white matter microstructure (i.e., fractional anisotropy and mean diffusivity), we examined: within- and between-sex variance, overlap between male and female distributions, inhomogeneity of variance via the Fligner-Killeen test, and an analysis of similarities (ANOSIM). For completeness, we examined these sex differences using both uncorrected (raw) brain estimates and residualized brain estimates after using mixed-effects modeling to account for age, pubertal development, socioeconomic status, race, ethnicity, MRI scanner manufacturer, and total brain volume, where applicable. Results The overlap between male and female distributions was universally greater than the difference (overlap coefficient range: 0.585 - 0.985) and the ratio of within-sex and between-sex differences was similar (ANOSIM R range: -0.001 - 0.117). All cortical and subcortical volumes showed significant inhomogeneity of variance, whereas a minority of brain regions showed significant sex differences in variance for cortical thickness, white matter volume, fractional anisotropy, and mean diffusivity. Inhomogeneity of variance was reduced after accounting for other sources of variance. Overlap coefficients were larger and ANOSIM R values were smaller for residualized outcomes, indicating greater within- and smaller between-sex differences once accounting for other covariates. Conclusions Reported sex differences in early adolescent human brain structure may be driven by disparities in variance, rather than binary, sex-based phenotypes. Contrary to the popular view of the brain as sexually dimorphic, we found more similarity than difference between sexes in all global and regional measurements of brain structure examined. This study builds upon previous findings illustrating the importance of considering variance when examining sex differences in brain structure.
Collapse
|
6
|
Pramanik S, Devi M H, Chakrabarty S, Paylar B, Pradhan A, Thaker M, Ayyadhury S, Manavalan A, Olsson PE, Pramanik G, Heese K. Microglia signaling in health and disease - Implications in sex-specific brain development and plasticity. Neurosci Biobehav Rev 2024; 165:105834. [PMID: 39084583 DOI: 10.1016/j.neubiorev.2024.105834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| | - Harini Devi M
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Saswata Chakrabarty
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Berkay Paylar
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Manisha Thaker
- Eurofins Lancaster Laboratories, Inc., 2425 New Holland Pike, Lancaster, PA 17601, USA
| | - Shamini Ayyadhury
- The Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Arulmani Manavalan
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600077, India
| | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro 70182, Sweden
| | - Gopal Pramanik
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand 835215, India.
| | - Klaus Heese
- Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 133791, the Republic of Korea.
| |
Collapse
|
7
|
Goetz SMM, Lucas T, Carré JM. Under the influence: exogenous testosterone influences men's cross-sex perceptions of sexual interest. Front Psychol 2024; 15:1425389. [PMID: 39315047 PMCID: PMC11418507 DOI: 10.3389/fpsyg.2024.1425389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024] Open
Abstract
The sexual misperception bias is a cognitive bias in which men tend to overestimate sexual interest from women, potentially shaped by evolutionary mating strategies. Testosterone, often linked to mating behaviors, might play a role in sustaining sexual overperceptions. To explore this possibility, we conducted a placebo-controlled study with 190 heterosexual men, administering either 11 mg of testosterone or a placebo. Participants interacted with an attractive female confederate, while naïve raters assessed the confederate's affiliative behaviors. Our findings suggest that exogenous testosterone did not broadly impact sexual overperception. However, we found that affiliative behavior from the confederate was positively correlated with perceived sexual interest among testosterone-treated, but not placebo-treated men. In addition, we found that this effect among testosterone-treated men was contingent on their self-perceived attractiveness. Specifically, the confederate's affiliative behaviors were positively correlated with perceived sexual interest, but only for testosterone-treated men with average or above average self-perceived attractiveness. Furthermore, our data revealed that men's tendency to project their own short-term and long-term mating interests increases as a function of self-perceived attractiveness, and this coupling is enhanced by testosterone for long-term interest. Taken together, these results suggest that testosterone may potentiate existing biases, particularly when sexual motivation is high, and bias perceptions of friendly behavior when engaging in cross-sex mindreading. This study adds to the understanding of the neuroendocrine bases of social cognition, suggesting that testosterone can affect men's perceptions of potential mates.
Collapse
Affiliation(s)
- Stefan M. M. Goetz
- Charles Stewart Mott Department of Public Health, College of Human Medicine, Michigan State University, Flint, MI, United States
| | - Todd Lucas
- Charles Stewart Mott Department of Public Health, College of Human Medicine, Michigan State University, Flint, MI, United States
| | - Justin M. Carré
- Laboratory of Social Neuroendocrinology, Department of Psychology, Nipissing University, North Bay, ON, Canada
| |
Collapse
|
8
|
Yeung J, DeYoung T, Spring S, de Guzman AE, Elder MW, Beauchamp A, Wong CS, Palmert MR, Lerch JP, Nieman BJ. Sex chromosomes and hormones independently influence healthy brain development but act similarly after cranial radiation. Proc Natl Acad Sci U S A 2024; 121:e2404042121. [PMID: 39207735 PMCID: PMC11388377 DOI: 10.1073/pnas.2404042121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
The course of normal development and response to pathology are strongly influenced by biological sex. For instance, female childhood cancer survivors who have undergone cranial radiation therapy (CRT) tend to display more pronounced cognitive deficits than their male counterparts. Sex effects can be the result of sex chromosome complement (XX vs. XY) and/or gonadal hormone influence. The contributions of each can be separated using the four-core genotype mouse model (FCG), where sex chromosome complement and gonadal sex are decoupled. While studies of FCG mice have evaluated brain differences in adulthood, it is still unclear how sex chromosome and sex hormone effects emerge through development in both healthy and pathological contexts. Our study utilizes longitudinal MRI with the FCG model to investigate sex effects in healthy development and after CRT in wildtype and immune-modified Ccl2-knockout mice. Our findings in normally developing mice reveal a relatively prominent chromosome effect prepubertally, compared to sex hormone effects which largely emerge later. Spatially, sex chromosome and hormone influences were independent of one another. After CRT in Ccl2-knockout mice, both male chromosomes and male hormones similarly improved brain outcomes but did so more separately than in combination. Our findings highlight the crucial role of sex chromosomes in early development and identify roles for sex chromosomes and hormones after CRT-induced inflammation, highlighting the influences of biological sex in both normal brain development and pathology.
Collapse
Affiliation(s)
- Jonas Yeung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Taylor DeYoung
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - A Elizabeth de Guzman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Rovereto TN 38068, Italy
| | - Madeline W Elder
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
| | - Antoine Beauchamp
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
| | - C Shun Wong
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, Toronto ON M4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, Toronto ON M5T 1P5, Canada
| | - Mark R Palmert
- Division of Endocrinology, The Hospital for Sick Children, University of Toronto, Toronto ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto ON M5S 1A8, Canada
- Department of Physiology, University of Toronto, Toronto ON M5S 1A8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Wellcome Centre for Integrative Neuroimaging, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, OXF OX3 9DU, United Kingdom
| | - Brian J Nieman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto ON M5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, Toronto ON M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto ON M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| |
Collapse
|
9
|
Luders E, Gaser C, Spencer D, Thankamony A, Hughes I, Srirangalingam U, Gleeson H, Hines M, Kurth F. Effects of Congenital Adrenal Hyperplasia (CAH) and Biological Sex on Brain Size. ANATOMIA (BASEL, SWITZERLAND) 2024; 3:155-162. [PMID: 39391581 PMCID: PMC11461354 DOI: 10.3390/anatomia3030012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Congenital Adrenal Hyperplasia (CAH) has been reported to involve structural alterations in some brain regions. However, it remains to be established whether there is also an impact on the size of the brain as a whole. Here, we compiled the largest CAH sample to date (n = 53), matched pair-wise to a control group (n = 53) on sex, age, and verbal intelligence. Using T1-weighted brain scans, we calculated intracranial volume (ICV) as well as total brain volume (TBV), which are both common estimates for brain size. The statistical analysis was performed using a general linear model assessing the effects of CAH (CAH vs. controls), sex (women vs. men), and any CAH-by-sex interaction. The outcomes were comparable for ICV and TBV, i.e., there was no significant main effect of CAH and no significant CAH-by-sex interaction. However, there was a significant main effect of sex, with larger ICVs and TBVs in men than in women. Our findings contribute to an understudied field of research exploring brain anatomy in CAH. In contrast to some existing studies suggesting a smaller brain size in CAH, we did not observe such an effect. In other words, ICV and TBV in women and men with CAH did not differ significantly from those in controls. Notwithstanding, we observed the well-known sex difference in brain size (12.69% for ICV and 12.50% for TBV), with larger volumes in men than in women, which is in agreement with the existing literature.
Collapse
Affiliation(s)
- Eileen Luders
- Department of Women’s and Children’s Health, Uppsala University, 75237 Uppsala, Sweden
- Swedish Collegium for Advanced Study (SCAS), 75238 Uppsala, Sweden
- School of Psychology, University of Auckland, Auckland 1010, New Zealand
- Laboratory of Neuro Imaging, School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Christian Gaser
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Department of Psychiatry and Psychotherapy, Jena University Hospital, 07747 Jena, Germany
| | - Debra Spencer
- Department of Psychology, University of Cambridge, Cambridge CB2 3RQ, UK
| | - Ajay Thankamony
- Department of Paediatrics, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
- Weston Centre for Paediatric Endocrinology & Diabetes, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Ieuan Hughes
- Department of Paediatrics, Addenbrooke’s Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Umasuthan Srirangalingam
- Department of Endocrinology and Diabetes, University College Hospital London, London NW1 2BU, UK
| | | | - Melissa Hines
- Department of Psychology, University of Cambridge, Cambridge CB2 3RQ, UK
| | - Florian Kurth
- School of Psychology, University of Auckland, Auckland 1010, New Zealand
- Department of Diagnostic and Interventional Radiology, Jena University Hospital, 07747 Jena, Germany
| |
Collapse
|
10
|
Pavlinek A, Adhya D, Tsompanidis A, Warrier V, Vernon AC, Lancaster M, Mill J, Srivastava DP, Baron-Cohen S. Using Organoids to Model Sex Differences in the Human Brain. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100343. [PMID: 39092139 PMCID: PMC11292257 DOI: 10.1016/j.bpsgos.2024.100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 08/04/2024] Open
Abstract
Sex differences are widespread during neurodevelopment and play a role in neuropsychiatric conditions such as autism, which is more prevalent in males than females. In humans, males have been shown to have larger brain volumes than females with development of the hippocampus and amygdala showing prominent sex differences. Mechanistically, sex steroids and sex chromosomes drive these differences in brain development, which seem to peak during prenatal and pubertal stages. Animal models have played a crucial role in understanding sex differences, but the study of human sex differences requires an experimental model that can recapitulate complex genetic traits. To fill this gap, human induced pluripotent stem cell-derived brain organoids are now being used to study how complex genetic traits influence prenatal brain development. For example, brain organoids from individuals with autism and individuals with X chromosome-linked Rett syndrome and fragile X syndrome have revealed prenatal differences in cell proliferation, a measure of brain volume differences, and excitatory-inhibitory imbalances. Brain organoids have also revealed increased neurogenesis of excitatory neurons due to androgens. However, despite growing interest in using brain organoids, several key challenges remain that affect its validity as a model system. In this review, we discuss how sex steroids and the sex chromosomes each contribute to sex differences in brain development. Then, we examine the role of X chromosome inactivation as a factor that drives sex differences. Finally, we discuss the combined challenges of modeling X chromosome inactivation and limitations of brain organoids that need to be taken into consideration when studying sex differences.
Collapse
Affiliation(s)
- Adam Pavlinek
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Dwaipayan Adhya
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Alex Tsompanidis
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Varun Warrier
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Anthony C. Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | | | - Jonathan Mill
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King’s College London, London, United Kingdom
| | - Simon Baron-Cohen
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
12
|
Wang Y, Riedstra B, Groothuis T. Effects of maternal androgens and their metabolite etiocholanolone on prenatal development in birds. J Exp Biol 2024; 227:jeb247205. [PMID: 39037123 PMCID: PMC11418167 DOI: 10.1242/jeb.247205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Offspring phenotypes can be affected by maternal testosterone and androstenedione (A4), which are considered a tool of mothers to adjust offspring to a fluctuating environment. Yet testosterone and A4 are very rapidly metabolized by developing avian embryos, suggesting that either the maternal testosterone and A4 have potent organizational effects on the embryos extremely early before being metabolized or it is the metabolites that evoke phenotypic variation in the offspring. One of the metabolites, etiocholanolone, increases substantially during early embryonic development and is a likely candidate for mediating maternal effects as it can promote erythropoiesis. To investigate and compare the effects of testosterone and A4 with the possible effects of etiocholanolone during prenatal embryonic development, we increased their levels in black-headed gull eggs (Larus ridibundus), and used sham-injected eggs as controls. This species usually has 3-egg clutches in which maternal androgen levels increase with the egg-laying sequence. We analysed embryonic heart rate, peri-hatching biometric traits, the ratio of white to red blood cells (W/R ratio) and bursa development. We found that testosterone and A4 treatment increased embryonic heart rate irrespective of egg-laying sequence and decreased bill length and W/R ratio, whereas etiocholanolone did not mimic these effects. Instead, etiocholanolone treatment decreased tarsus length and brain mass. Our finding that etiocholanolone does not mimic the effects induced by testosterone and A4 suggests that the embryonic metabolism of maternal testosterone and A4 can potentially diversify the function of these maternal androgens.
Collapse
Affiliation(s)
- Yuqi Wang
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| | - Bernd Riedstra
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| | - Ton Groothuis
- University of Groningen, Groningen Institute for Evolutionary Life Sciences, 9700 AB Groningen, The Netherlands
| |
Collapse
|
13
|
Sheng JA, Tobet SA. Maternal immune activation with toll-like receptor 7 agonist during mid-gestation alters juvenile and adult developmental milestones and behavior. J Neuroendocrinol 2024; 36:e13417. [PMID: 38822791 PMCID: PMC11296912 DOI: 10.1111/jne.13417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/07/2024] [Accepted: 05/15/2024] [Indexed: 06/03/2024]
Abstract
Infections during pregnancy are associated with increased risk for adult neuropsychiatric disease, such as major depressive disorder, schizophrenia, and autism spectrum disorder. In mouse models of maternal immune activation (MIA), different toll-like receptors (TLRs) are stimulated to initiate inflammatory responses in mother and fetus. The goal of this study was to determine sex-dependent aspects of MIA using a TLR7/8 agonist, Resiquimod (RQ), on neurodevelopment. RQ was administered to timed-pregnant mice on embryonic day (E) 12.5. At E15, maternal/fetal plasma cytokines were measured by enzyme-linked immunosorbent assay (ELISA). Maternal cytokines interleukin (IL)-6 and IL-10 were higher while tumor necrosis factor (TNF)-α and IL-17 were lower in pregnant dams exposed to RQ. Fetal cytokines (E15) were altered at the same timepoint with fetal plasma IL-6 and IL-17 greater after RQ compared to vehicle, while IL-10 and TNF-α were higher in male fetuses but not female. Other timed-pregnant dams were allowed to give birth. MIA with RQ did not alter the female to male ratio of offspring born per litter. Body weights were reduced significantly in both sexes at birth, and over the next 5 weeks. Offspring from RQ-injected mothers opened their eyes 5 days later than controls. Similarly, female offspring from RQ-injected mothers exhibited pubertal delay based on vaginal opening 2-3 days later than control females. On the behavioral side, juvenile and adult male and female MIA offspring exhibited less social-like behavior in a social interaction test. Anhedonia-like behavior was greater in MIA adult female mice. This study provides support for sex-dependent influences of fetal antecedents for altered brain development and behavioral outputs that could be indicative of increased susceptibility for adult disorders through immune mechanisms. Future studies are needed to determine neural cellular and molecular mechanisms for such programming effects.
Collapse
Affiliation(s)
| | - Stuart A. Tobet
- Biomedical Sciences, Colorado State University, Fort Collins, CO
- Department of Psychiatry, Mass General Hospital, Harvard Medical School, Boston, MA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO
- Innovation Center on Sex Differences in Medicine, Mass General Hospital
| |
Collapse
|
14
|
Kissel LT, Pochareddy S, An JY, Sestan N, Sanders SJ, Wang X, Werling DM. Sex-Differential Gene Expression in Developing Human Cortex and Its Intersection With Autism Risk Pathways. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100321. [PMID: 38957312 PMCID: PMC11217612 DOI: 10.1016/j.bpsgos.2024.100321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 07/04/2024] Open
Abstract
Background Sex-differential biology may contribute to the consistently male-biased prevalence of autism spectrum disorder (ASD). Gene expression differences between males and females in the brain can indicate possible molecular and cellular mechanisms involved, although transcriptomic sex differences during human prenatal cortical development have been incompletely characterized, primarily due to small sample sizes. Methods We performed a meta-analysis of sex-differential expression and co-expression network analysis in 2 independent bulk RNA sequencing datasets generated from cortex of 273 prenatal donors without known neuropsychiatric disorders. To assess the intersection between neurotypical sex differences and neuropsychiatric disorder biology, we tested for enrichment of ASD-associated risk genes and expression changes, neuropsychiatric disorder risk genes, and cell type markers within identified sex-differentially expressed genes (sex-DEGs) and sex-differential co-expression modules. Results We identified 101 significant sex-DEGs, including Y-chromosome genes, genes impacted by X-chromosome inactivation, and autosomal genes. Known ASD risk genes, implicated by either common or rare variants, did not preferentially overlap with sex-DEGs. We identified 1 male-specific co-expression module enriched for immune signaling that is unique to 1 input dataset. Conclusions Sex-differential gene expression is limited in prenatal human cortex tissue, although meta-analysis of large datasets allows for the identification of sex-DEGs, including autosomal genes that encode proteins involved in neural development. Lack of sex-DEG overlap with ASD risk genes in the prenatal cortex suggests that sex-differential modulation of ASD symptoms may occur in other brain regions, at other developmental stages, or in specific cell types, or may involve mechanisms that act downstream from mutation-carrying genes.
Collapse
Affiliation(s)
- Lee T. Kissel
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sirisha Pochareddy
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- Transdisciplinary Major in Learning Health Systems, Department of Healthcare Sciences, Graduate School, Korea University, Seoul, Republic of Korea
- BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, Connecticut
| | - Stephan J. Sanders
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Department of Psychiatry and Behavioral Sciences, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California
| | - Xuran Wang
- Seaver Autism Center for Research and Treatment, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Donna M. Werling
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
15
|
Paylar B, Pramanik S, Bezabhe YH, Olsson PE. Temporal sex specific brain gene expression pattern during early rat embryonic development. Front Cell Dev Biol 2024; 12:1343800. [PMID: 38961864 PMCID: PMC11219815 DOI: 10.3389/fcell.2024.1343800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 06/05/2024] [Indexed: 07/05/2024] Open
Abstract
Background: The classical concept of brain sex differentiation suggests that steroid hormones released from the gonads program male and female brains differently. However, several studies indicate that steroid hormones are not the only determinant of brain sex differentiation and that genetic differences could also be involved. Methods: In this study, we have performed RNA sequencing of rat brains at embryonic days 12 (E12), E13, and E14. The aim was to identify differentially expressed genes between male and female rat brains during early development. Results: Analysis of genes expressed with the highest sex differences showed that Xist was highly expressed in females having XX genotype with an increasing expression over time. Analysis of genes expressed with the highest male expression identified three early genes, Sry2, Eif2s3y, and Ddx3y. Discussion: The observed sex-specific expression of genes at early development confirms that the rat brain is sexually dimorphic prior to gonadal action on the brain and identifies Sry2 and Eif2s3y as early genes contributing to male brain development.
Collapse
Affiliation(s)
| | | | | | - Per-Erik Olsson
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
16
|
Casto KV, Cohen DJ, Akinola M, Mehta PH. Testosterone, gender identity and gender-stereotyped personality attributes. Horm Behav 2024; 162:105540. [PMID: 38652981 DOI: 10.1016/j.yhbeh.2024.105540] [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: 10/30/2023] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/25/2024]
Abstract
Sex/gender differences in personality associated with gender stereotyped behavior are widely studied in psychology yet remain a subject of ongoing debate. Exposure to testosterone during developmental periods is considered to be a primary mediator of many sex/gender differences in behavior. Extensions of this research has led to both lay beliefs and initial research about individual differences in basal testosterone in adulthood relating to "masculine" personality. In this study, we explored the relationships between testosterone, gender identity, and gender stereotyped personality attributes in a sample of over 400 university students (65 % female assigned at birth). Participants provided ratings of their self-perceived masculinity and femininity, resulting in a continuous measure of gender identity, and a set of agentic and communal personality attributes. A saliva sample was also provided for assay of basal testosterone. Results showed no compelling evidence that basal testosterone correlates with gender-stereotyped personality attributes or explains the relationship between sex/gender identity and these attributes, across, within, or covarying out sex assigned at birth. Contributing to a more gender diverse approach to assessing sex/gender relationships with personality and testosterone, our continuous measure of self-perceived masculinity and femininity predicted additional variance in personality beyond binary sex and showed some preliminary but weak relationships with testosterone. Results from this study cast doubt on the activational testosterone-masculinity hypothesis for explaining sex differences in gender stereotyped traits and within-sex/gender variation in attributes associated with agency and communality.
Collapse
Affiliation(s)
- Kathleen V Casto
- Kent State University, Department of Psychological Sciences, United States of America.
| | - Dale J Cohen
- University of North Carolina Wilmington, Department of Psychology, United States of America
| | - Modupe Akinola
- Columbia University, Columbia Business School, United States of America
| | - Pranjal H Mehta
- University College London, Department of Experimental Psychology, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
17
|
Inoue S. Hormonal and circuit mechanisms controlling female sexual behavior. Front Neural Circuits 2024; 18:1409349. [PMID: 38752168 PMCID: PMC11094328 DOI: 10.3389/fncir.2024.1409349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/15/2024] [Indexed: 05/18/2024] Open
Abstract
Sexual behavior is crucial for reproduction in many animals. In many vertebrates, females exhibit sexual behavior only during a brief period surrounding ovulation. Over the decades, studies have identified the roles of ovarian sex hormones, which peak in levels around the time of ovulation, and the critical brain regions involved in the regulation of female sexual behavior. Modern technical innovations have enabled a deeper understanding of the neural circuit mechanisms controlling this behavior. In this review, I summarize our current knowledge and discuss the neural circuit mechanisms by which female sexual behavior occurs in association with the ovulatory phase of their cycle.
Collapse
Affiliation(s)
- Sayaka Inoue
- Department of Psychiatry, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
18
|
Kung KTF, Louie K, Spencer D, Hines M. Prenatal androgen exposure and sex-typical play behaviour: A meta-analysis of classic congenital adrenal hyperplasia studies. Neurosci Biobehav Rev 2024; 159:105616. [PMID: 38447820 DOI: 10.1016/j.neubiorev.2024.105616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
Thousands of non-human mammal experiments have demonstrated that early androgen exposure exerts long-lasting effects on neurobehavioural sexual differentiation. In humans, females with classic congenital adrenal hyperplasia (CAH) are exposed to unusually high concentrations of androgens prenatally, whereas prenatal concentrations of androgens in males with CAH are largely normal. The current meta-analysis included 20 independent samples and employed multi-level meta-analytic models. Consistently across all 7 male-typical and female-typical play outcomes, in the expected directions, the present study found significant and large average differences between control males and control females (gs = 0.83-2.78) as well as between females with CAH and control females (gs = 0.95-1.08), but differences between males with CAH and control males were mostly negligible and were non-significant for 6 of the 7 outcomes (gs = 0.04-0.27). These meta-analytic findings suggest that prenatal androgen exposure masculinises and defeminises play behaviour in humans. Broader implications in relation to sex chromosomes, brain development, oestrogens, socio-cognitive influences, other aspects of sex-related behavioural development, and gender nonconformity are discussed.
Collapse
Affiliation(s)
- Karson T F Kung
- Department of Psychology, Jockey Club Tower, Centennial Campus, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region.
| | - Krisya Louie
- Department of Psychology, Jockey Club Tower, Centennial Campus, University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region
| | - Debra Spencer
- Department of Psychology, University of Cambridge, Free School Lane, Cambridge CB2 3RQ, United Kingdom
| | - Melissa Hines
- Department of Psychology, University of Cambridge, Free School Lane, Cambridge CB2 3RQ, United Kingdom
| |
Collapse
|
19
|
García-Fernández L, Chavira DR, Hoffman K, González-Mariscal G. Effects of intrauterine position during gestation on specific endocrine and behavioral parameters that impact reproduction in domestic rabbits. Horm Behav 2024; 160:105503. [PMID: 38342021 DOI: 10.1016/j.yhbeh.2024.105503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Prior studies from others, performed in a different breed, reported that doe rabbits developing between two male siblings (2 M) during gestation display characteristics indicative of masculinization: larger anogenital distance (AGD), larger submandibular glands, and higher chinning frequency than females with zero (0 M) or one (1 M) contiguous brothers. Similar effects are provoked by injecting androgens to the pregnant doe suggesting that prenatal androgen exposure masculinizes female embryos. To further understand the scope of such masculinization we compared 0 M, 1 M, and 2 M females regarding behavioral, neuroendocrine, and somatic parameters, related or not to reproduction. IUP did not impact: body weight, sexual receptivity, mating-induced LH secretion, maternal nest-building, litter size, or milk output. At puberty: a) chinning frequency was: 0 M and males>1 M and 2 M; b) ambulation in open field was lowest in 1 M females and males. IUP effects on AGD were significant only on postnatal day 1: 0 M, 1 M, and males>2 M, in contrast to earlier study. Willingness to nurse at delivery was less frequent in 2 M than in 1 M and 0 M does and correlated with nursing occurrence across lactation. Does that did not nurse at parturition delivered fewer kits/min than those that nursed then, regardless of IUP. The duration of nursing bouts across lactation was significantly longer in the1 M and 2 M does that showed this behavior on postpartum days 1-20. Our findings indicate that IUP is associated with alterations in specific aspects of postpartum maternal behavior.
Collapse
Affiliation(s)
- Lizet García-Fernández
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Mexico
| | - David R Chavira
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Mexico
| | - Kurt Hoffman
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, Mexico
| | | |
Collapse
|
20
|
Abstract
In recent years, the impact of prenatal sound on development, notably for programming individual phenotypes for postnatal conditions, has increasingly been revealed. However, the mechanisms through which sound affects physiology and development remain mostly unexplored. Here, I gather evidence from neurobiology, developmental biology, cellular biology and bioacoustics to identify the most plausible modes of action of sound on developing embryos. First, revealing often-unsuspected plasticity, I discuss how prenatal sound may shape auditory system development and determine individuals' later capacity to receive acoustic information. I also consider the impact of hormones, including thyroid hormones, glucocorticoids and androgen, on auditory plasticity. Second, I review what is known about sound transduction to other - non-auditory - brain regions, and its potential to input on classical developmental programming pathways. Namely, the auditory pathway has direct anatomical and functional connectivity to the hippocampus, amygdala and/or hypothalamus, in mammals, birds and anurans. Sound can thus trigger both immediate and delayed responses in these limbic regions, which are specific to the acoustic stimulus and its biological relevance. Third, beyond the brain, I briefly consider the possibility for sound to directly affect cellular functioning, based on evidence in earless organisms (e.g. plants) and cell cultures. Together, the multi-disciplinary evidence gathered here shows that the brain is wired to allow multiple physiological and developmental effects of sound. Overall, there are many unexplored, but possible, pathways for sound to impact even primitive or immature organisms. Throughout, I identify the most promising research avenues for unravelling the processes of acoustic developmental programming.
Collapse
Affiliation(s)
- Mylene M Mariette
- Doñana Biological Station EBD-CSIC, 41092 Seville, Spain
- School of Life and Environmental Sciences, Deakin University, Geelong, VIC 3216, Australia
| |
Collapse
|
21
|
Carrillo B, Fernandez-Garcia JM, García-Úbeda R, Grassi D, Primo U, Blanco N, Ballesta A, Arevalo MA, Collado P, Pinos H. Neonatal inhibition of androgen activity alters the programming of body weight and orexinergic peptides differentially in male and female rats. Brain Res Bull 2024; 208:110898. [PMID: 38360152 DOI: 10.1016/j.brainresbull.2024.110898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
The involvement of androgens in the regulation of energy metabolism has been demonstrated. The main objective of the present research was to study the involvement of androgens in both the programming of energy metabolism and the regulatory peptides associated with feeding. For this purpose, androgen receptors and the main metabolic pathways of testosterone were inhibited during the first five days of postnatal life in male and female Wistar rats. Pups received a daily s.c. injection from the day of birth, postnatal day (P) 1, to P5 of Flutamide (a competitive inhibitor of androgen receptors), Letrozole (an aromatase inhibitor), Finasteride (a 5-alpha-reductase inhibitor) or vehicle. Body weight, food intake and fat pads were measured. Moreover, hypothalamic Agouti-related peptide (AgRP), neuropeptide Y (NPY), orexin, and proopiomelanocortin (POMC) were analyzed by quantitative real-time polymerase chain reaction assay. The inhibition of androgenic activity during the first five days of life produced a significant decrease in body weight in females at P90 but did not affect this parameter in males. Moreover, the inhibition of aromatase decreased hypothalamic AgRP mRNA levels in males while the inhibition of 5α-reductase decreased hypothalamic AgRP and orexin mRNA levels in female rats. Finally, food intake and visceral fat, but not subcutaneous fat, were affected in both males and females depending on which testosterone metabolic pathway was inhibited. Our results highlight the differential involvement of androgens in the programming of energy metabolism as well as the AgRP and orexin systems during development in male and female rats.
Collapse
Affiliation(s)
- Beatriz Carrillo
- Department of Psychobiology, National University of Distance Education, Madrid, Spain; University Institute of Research-UNED-Institute of Health Carlos III (IMIENS), Madrid, Spain
| | - Jose Manuel Fernandez-Garcia
- University Institute of Research-UNED-Institute of Health Carlos III (IMIENS), Madrid, Spain; Faculty of Psychology, Universidad Villanueva Madrid, Madrid, Spain
| | - Rocío García-Úbeda
- Department of Psychobiology, National University of Distance Education, Madrid, Spain
| | - Daniela Grassi
- Department of Anatomy, Histology and Neuroscience, Autonomous University of Madrid, Madrid, Spain
| | - Ulises Primo
- Department of Psychobiology, National University of Distance Education, Madrid, Spain
| | - Noemí Blanco
- Department of Psychobiology, National University of Distance Education, Madrid, Spain; University Institute of Research-UNED-Institute of Health Carlos III (IMIENS), Madrid, Spain
| | - Antonio Ballesta
- Department of Psychobiology, Centro de Enseñanza Superior Cardenal Cisneros, Spain
| | - Maria Angeles Arevalo
- Neuroactive Steroids Lab, Cajal Institute, CSIC, Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Paloma Collado
- Department of Psychobiology, National University of Distance Education, Madrid, Spain; University Institute of Research-UNED-Institute of Health Carlos III (IMIENS), Madrid, Spain
| | - Helena Pinos
- Department of Psychobiology, National University of Distance Education, Madrid, Spain; University Institute of Research-UNED-Institute of Health Carlos III (IMIENS), Madrid, Spain.
| |
Collapse
|
22
|
Veyrunes F, Perez J, Heitzmann LD, Saunders PA, Givalois L. Hormone profiles of the African pygmy mouse Mus minutoides, a species with XY female sex reversal. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:130-137. [PMID: 38059664 DOI: 10.1002/jez.2767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 12/08/2023]
Abstract
In mammals, most sex differences in phenotype are controlled by gonadal hormones, but recent work on transgenic mice has shown that sex chromosomes can have a direct influence on sex-specific behaviors. In this study, we take advantage of the naturally occurring sex reversal in a mouse species, Mus minutoides, to investigate for the first time the relationship between sex chromosomes, hormones, and behaviors in a wild species. In this model, a feminizing variant of the X chromosome, named X*, produces three types of females with different sex chromosome complements (XX, XX*, and X*Y), associated with alternative behavioral phenotypes, while all males are XY. We thus compared the levels of three major circulating steroid hormones (testosterone, corticosterone, and estradiol) in the four sex genotypes to disentangle the influence of sex chromosomes and sex hormones on behavior. First, we did not find any difference in testosterone levels in the three female genotypes, although X*Y females are notoriously more aggressive. Second, in agreement with their lower anxiety-related behaviors, X*Y females and XY males display lower baseline corticosterone concentration than XX and XX* females. Instead of a direct hormonal influence, this result rather suggests that sex chromosomes may have an impact on the baseline corticosterone level, which in turn may influence behaviors. Third, estradiol concentrations do not explain the enhanced reproductive performance and maternal care behavior of the X*Y females compared to the XX and XX* females. Overall, this study highlights that most of the behaviors varying along with sex chromosome complement of this species are more likely driven by genetic factors rather than steroid hormone concentrations.
Collapse
Affiliation(s)
- Frederic Veyrunes
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Julie Perez
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Louise D Heitzmann
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Paul A Saunders
- ISEM, Institut des Sciences de l'Evolution de Montpellier UMR 5554, CNRS, Université Montpellier, IRD, Montpellier, France
| | - Laurent Givalois
- MMDN, Molecular Mechanisms in Neurodegenerative Dementia Laboratory, Université Montpellier, EPHE-PSL, INSERM U1198, Montpellier, France
- Department of Psychiatry and Neurosciences, CR-CHUQ, Faculty of Medicine, Laval University, Québec City, Canada
- CNRS, Paris, France
| |
Collapse
|
23
|
Christians JK, Reue K. The role of gonadal hormones and sex chromosomes in sex-dependent effects of early nutrition on metabolic health. Front Endocrinol (Lausanne) 2023; 14:1304050. [PMID: 38189044 PMCID: PMC10770830 DOI: 10.3389/fendo.2023.1304050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Early-life conditions such as prenatal nutrition can have long-term effects on metabolic health, and these effects may differ between males and females. Understanding the biological mechanisms underlying sex differences in the response to early-life environment will improve interventions, but few such mechanisms have been identified, and there is no overall framework for understanding sex differences. Biological sex differences may be due to chromosomal sex, gonadal sex, or interactions between the two. This review describes approaches to distinguish between the roles of chromosomal and gonadal sex, and summarizes findings regarding sex differences in metabolism. The Four Core Genotypes (FCG) mouse model allows dissociation of the sex chromosome genotype from gonadal type, whereas the XY* mouse model can be used to distinguish effects of X chromosome dosage vs the presence of the Y chromosome. Gonadectomy can be used to distinguish between organizational (permanent) and activational (reversible) effects of sex hormones. Baseline sex differences in a variety of metabolic traits are influenced by both activational and organizational effects of gonadal hormones, as well as sex chromosome complement. Thus far, these approaches have not been widely applied to examine sex-dependent effects of prenatal conditions, although a number of studies have found activational effects of estradiol to be protective against the development of hypertension following early-life adversity. Genes that escape X chromosome inactivation (XCI), such as Kdm5c, contribute to baseline sex-differences in metabolism, while Ogt, another XCI escapee, leads to sex-dependent responses to prenatal maternal stress. Genome-wide approaches to the study of sex differences include mapping genetic loci influencing metabolic traits in a sex-dependent manner. Seeking enrichment for binding sites of hormone receptors among genes showing sexually-dimorphic expression can elucidate the relative roles of hormones. Using the approaches described herein to identify mechanisms underlying sex-dependent effects of early nutrition on metabolic health may enable the identification of fundamental mechanisms and potential interventions.
Collapse
Affiliation(s)
- Julian K. Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
- Women’s Health Research Institute, BC Women’s Hospital and Health Centre, Vancouver, BC, Canada
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
24
|
Lischinsky JE, Yin L, Shi C, Prakash N, Burke J, Shekaran G, Grba M, Corbin JG, Lin D. Transcriptionally defined amygdala subpopulations play distinct roles in innate social behaviors. Nat Neurosci 2023; 26:2131-2146. [PMID: 37946049 PMCID: PMC10689240 DOI: 10.1038/s41593-023-01475-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
Social behaviors are innate and supported by dedicated neural circuits, but the molecular identities of these circuits and how they are established developmentally and shaped by experience remain unclear. Here we show that medial amygdala (MeA) cells originating from two embryonically parcellated developmental lineages have distinct response patterns and functions in social behavior in male mice. MeA cells expressing the transcription factor Foxp2 (MeAFoxp2) are specialized for processing male conspecific cues and are essential for adult inter-male aggression. By contrast, MeA cells derived from the Dbx1 lineage (MeADbx1) respond broadly to social cues, respond strongly during ejaculation and are not essential for male aggression. Furthermore, MeAFoxp2 and MeADbx1 cells show differential anatomical and functional connectivity. Altogether, our results suggest a developmentally hardwired aggression circuit at the MeA level and a lineage-based circuit organization by which a cell's embryonic transcription factor profile determines its social information representation and behavioral relevance during adulthood.
Collapse
Affiliation(s)
- Julieta E Lischinsky
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
| | - Luping Yin
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Chenxi Shi
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Hunter College, New York, NY, USA
| | - Nandkishore Prakash
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Jared Burke
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Govind Shekaran
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Maria Grba
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Dayu Lin
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Ji X, Li X, Wang L, Liu S, Jiang X, Pan Y. Asexuality in Drosophila juvenile males is organizational and independent of juvenile hormone. EMBO Rep 2023; 24:e56898. [PMID: 37530648 PMCID: PMC10561357 DOI: 10.15252/embr.202356898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/26/2023] [Accepted: 07/14/2023] [Indexed: 08/03/2023] Open
Abstract
Sexuality is generally prevented in newborns and arises with organizational rewiring of neural circuitry and optimization of fitness for reproduction competition. Recent studies reported that sex circuitry in Drosophila melanogaster is developed in juvenile males but functionally inhibited by juvenile hormone (JH). Here, we find that the fly sex circuitry, mainly expressing the male-specific fruitless (fruM ) and/or doublesex (dsx), is organizationally undeveloped and functionally inoperative in juvenile males. Artificially activating all fruM neurons induces substantial courtship in solitary adult males but not in juvenile males. Synaptic transmissions between major courtship regulators and all dsx neurons are strong in adult males but either weak or undetectable in juvenile males. We further find that JH does not inhibit male courtship in juvenile males but instead promotes courtship robustness in adult males. Our results indicate that the transition to sexuality from juvenile to adult flies requires organizational rewiring of neural circuitry.
Collapse
Affiliation(s)
- Xiaoxiao Ji
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and TechnologySoutheast UniversityNanjingChina
| | - Xiaolong Li
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and TechnologySoutheast UniversityNanjingChina
| | - Lin Wang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and TechnologySoutheast UniversityNanjingChina
| | - Suning Liu
- Guangmeiyuan R&D Center, Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied TechnologySouth China Normal UniversityMeizhouChina
| | - Xinyu Jiang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and TechnologySoutheast UniversityNanjingChina
| | - Yufeng Pan
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and TechnologySoutheast UniversityNanjingChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
26
|
Abstract
Unreliable protein-based tools are impeding sex-based research.
Collapse
Affiliation(s)
- Bradley D Gelfand
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
27
|
Bobotis BC, Braniff O, Gargus M, Akinluyi ET, Awogbindin IO, Tremblay MÈ. Sex differences of microglia in the healthy brain from embryonic development to adulthood and across lifestyle influences. Brain Res Bull 2023; 202:110752. [PMID: 37652267 DOI: 10.1016/j.brainresbull.2023.110752] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Microglia, the central nervous system innate immune cells, play a critical role in maintaining a homeostatic environment in the brain throughout life. These cells exhibit an impressive range of functions and characteristics that help to ensure proper functioning of the brain. Notably, microglia can present differences in their genetic and physical traits, which can be influenced by a range of factors, including age, environmental exposures, disease, and sex. Remarkably, microglia have been found to express receptors for sex hormones, suggesting that these hormones may play a role in modulating microglial behavior and potentially contribute to sex differences. Additionally, sex-chromosomal factors were shown to impact microglial genetics and functioning. In this review, we will examine how microglial responses in homeostasis are impacted by their interaction with sex hormones and sex chromosomes. Specifically, our investigation will focus on examining this interaction from embryonic development to adulthood, and the influence of lifestyle elements on various microglial features, including density and distribution, morphology, transcriptome, and proteome.
Collapse
Affiliation(s)
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Ifeoluwa Oluleke Awogbindin
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neuroimmunology Group, Molecular Drug Metabolism and Toxicology Laboratory, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec, QC, Canada; Department of Molecular Medicine, Université Laval, Québec, QC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
| |
Collapse
|
28
|
Little KM, Kosten TA. Focus on fentanyl in females: Sex and gender differences in the physiological and behavioral effects of fentanyl. Front Neuroendocrinol 2023; 71:101096. [PMID: 37597668 DOI: 10.1016/j.yfrne.2023.101096] [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: 03/30/2023] [Revised: 08/08/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
The prevalence of opioid use disorder and overdose continues to harm the U.S. population and is further exacerbated by the use of the synthetic opioid, fentanyl, and its analogs. Gender differences in the effects of fentanyl are not well understood. The present article reviews evidence for gender and sex differences in the physiological and behavioral effects of fentanyl in humans and animals. Biological sex seems to be a foundational driver in addiction vulnerability and affects mechanisms related to opioid use including fentanyl. Fentanyl has distinct pharmacodynamics and enhanced efficacy relative to other opioids that highlights the need to investigate how females may be uniquely altered by its use. Behavioral and physiological responses to fentanyl are found to differ by sex and gender in many cases, including outputs like affective symptoms, analgesia, tolerance, and withdrawal emphasizing the need for further research about the role of biological sex on fentanyl use.
Collapse
Affiliation(s)
- Kaitlyn M Little
- Department of Psychology, University of Houston, Houston, TX, United States
| | - Therese A Kosten
- Department of Psychology, University of Houston, Houston, TX, United States.
| |
Collapse
|
29
|
Sethuram S, Raymond S, Wang C, Barrett ES, Bush NR, Nguyen R, Sathyanarayana S, Swan SH, Evans SF. Early prenatal sex steroids and sex-typed play behavior at 4 years of age. Psychoneuroendocrinology 2023; 156:106288. [PMID: 37480735 DOI: 10.1016/j.psyneuen.2023.106288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/27/2023] [Accepted: 05/02/2023] [Indexed: 07/24/2023]
Abstract
During pregnancy, estrogens and testosterone influence brain development, resulting in sex-typical behavioral phenotypes. Prenatal testosterone exposure is associated with more male-typical behaviors in rodents, monkeys, and humans; however, few studies have examined the relationship between maternal sex hormones within the normal range and sex-dimorphic behaviors. In this study, we examined associations between prenatal estrogens and testosterone and sex-typical play in The Infant Development and the Environment Study (TIDES), a multicenter pregnancy cohort. We collected prenatal serum during the first trimester (mean=11.1 ± 2.6 weeks) and assessed child play behavior using the maternally completed Pre-School Activities Inventory (PSAI) at a mean age of 4.5 ± 0.3 years. This analysis includes mother-child pairs with complete data on hormones, play behavior, and covariates (n = 192 boys and 207 girls). No associations were seen between testosterone and PSAI scores in boys or girls or between estrogens and PSAI scores in boys. In girls, we observed an inverse relationship between feminine PSAI scores and both estradiol (E2) and estriol (E3) in multivariable linear regression analyses (E2: -0.11 [95% CI -0.20, -0.02]; E3: -0.44 [95% CI -0.83,-0.04]). Because the relationship between sex hormones and PSAI scores appeared nonlinear, we fit piecewise regression models to better fit the data and identify inflection points (point at which there is a significant change in slope). Piecewise regression analyses yielded inverse associations between masculine PSAI scores and estrone (E1) at values of E1 > 1340 pg/mL and E2 at values of E2 > 2870 pg/mL in girls. Further studies are needed to better understand the role of prenatal sex steroids on sexually dimorphic behavior.
Collapse
Affiliation(s)
- Swathi Sethuram
- Department of Pediatric Endocrinology, Massachusetts General Hospital, Boston, MA, USA.
| | - Samantha Raymond
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Wang
- Clinical and Translational Science Institute, The Lundquist Institute and Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Nicole R Bush
- Departments of Psychiatry and Pediatrics, Center for Health and Community, University of California, San Francisco, CA, USA
| | - Ruby Nguyen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, USA
| | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Shanna H Swan
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah Felice Evans
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
30
|
Ziemka-Nalecz M, Pawelec P, Ziabska K, Zalewska T. Sex Differences in Brain Disorders. Int J Mol Sci 2023; 24:14571. [PMID: 37834018 PMCID: PMC10572175 DOI: 10.3390/ijms241914571] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
A remarkable feature of the brain is its sexual dimorphism. Sexual dimorphism in brain structure and function is associated with clinical implications documented previously in healthy individuals but also in those who suffer from various brain disorders. Sex-based differences concerning some features such as the risk, prevalence, age of onset, and symptomatology have been confirmed in a range of neurological and neuropsychiatric diseases. The mechanisms responsible for the establishment of sex-based differences between men and women are not fully understood. The present paper provides up-to-date data on sex-related dissimilarities observed in brain disorders and highlights the most relevant features that differ between males and females. The topic is very important as the recognition of disparities between the sexes might allow for the identification of therapeutic targets and pharmacological approaches for intractable neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | | | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106 Warsaw, Poland; (M.Z.-N.); (P.P.); (K.Z.)
| |
Collapse
|
31
|
Konadu ME, Reed MB, Kaufmann U, Handschuh PA, Spurny-Dworak B, Klöbl M, Schmidt C, Godber, Godbersen M, Briem E, Seiger R, Baldinger-Melich P, Kranz GS, Lanzenberger R, Spies M. Changes to hypothalamic volume and associated subunits during gender-affirming hormone therapy. J Psychiatry Neurosci 2023; 48:E369-E375. [PMID: 37751919 PMCID: PMC10521920 DOI: 10.1503/jpn.230017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/30/2023] [Accepted: 08/01/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Among its pleiotropic properties, gender-affirming hormone therapy (GHT) affects regional brain volumes. The hypothalamus, which regulates neuroendocrine function and associated emotional and cognitive processes, is an intuitive target for probing GHT effects. We sought to assess changes to hypothalamus and hypothalamic subunit volumes after GHT, thereby honouring the region's anatomical and functional heterogeneity. METHODS Individuals with gender dysphoria and cisgender controls underwent 2 MRI measurements, with a median interval of 145 days (interquartile range [IQR] 128.25-169.75 d, mean 164.94 d) between the first and second MRI. Transgender women (TW) and transgender men (TM) underwent the first MRI before GHT and the second MRI after approximately 4.5 months of GHT, which comprised estrogen and anti-androgen therapy in TW or testosterone therapy in TM. Hypothalamic volumes were segmented using FreeSurfer, and effects of GHT were tested using repeated-measures analysis of covariance. RESULTS The final sample included 106 participants: 38 TM, 15 TW, 32 cisgender women (CW) and 21 cisgender men (CM). Our analyses revealed group × time interaction effects for total, left and right hypothalamus volume, and for several subunits (left and right inferior tubular, left superior tubular, right anterior inferior, right anterior superior, all p corr < 0.01). In TW, volumes decreased between the first and second MRI in these regions (all p corr ≤ 0.01), and the change from the first to second MRI in TW differed significantly from that in CM and CW in several subunits (p corr < 0.05). LIMITATIONS We did not address the influence of transition-related psychological and behavioural changes. CONCLUSION Our results suggest a subunit-specific effect of GHT on hypothalamus volumes in TW. This finding is in accordance with previous reports of positive and negative effects of androgens and estrogens, respectively, on cerebral volumes.
Collapse
Affiliation(s)
- Melisande E Konadu
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Murray B Reed
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Ulrike Kaufmann
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Patricia A Handschuh
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Benjamin Spurny-Dworak
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Manfred Klöbl
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Clemens Schmidt
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Godber
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - M Godbersen
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Elisa Briem
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - René Seiger
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Pia Baldinger-Melich
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Georg S Kranz
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Rupert Lanzenberger
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| | - Marie Spies
- From the Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Kranz, Lanzenberger, Spies); the Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria (Konadu, Reed, Handschuh, Spurny-Dworak, Klöbl, Schmidt, Godbersen, Briem, Seiger, Baldinger-Melich, Lanzenberger, Spies); the Department of Obstetrics and Gynecology, Medical University of Vienna, Austria (Kaufmann); the Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong (Kranz)
| |
Collapse
|
32
|
Kheloui S, Jacmin-Park S, Larocque O, Kerr P, Rossi M, Cartier L, Juster RP. Sex/gender differences in cognitive abilities. Neurosci Biobehav Rev 2023; 152:105333. [PMID: 37517542 DOI: 10.1016/j.neubiorev.2023.105333] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 07/09/2023] [Accepted: 07/27/2023] [Indexed: 08/01/2023]
Abstract
Sex/gender differences in cognitive sciences are riddled by conflicting perspectives. At the center of debates are clinical, social, and political perspectives. Front and center, evolutionary and biological perspectives have often focused on 'nature' arguments, while feminist and constructivist views have often focused on 'nurture arguments regarding cognitive sex differences. In the current narrative review, we provide a comprehensive overview regarding the origins and historical advancement of these debates while providing a summary of the results in the field of sexually polymorphic cognition. In so doing, we attempt to highlight the importance of using transdisciplinary perspectives which help bridge disciplines together to provide a refined understanding the specific factors that drive sex differences a gender diversity in cognitive abilities. To summarize, biological sex (e.g., birth-assigned sex, sex hormones), socio-cultural gender (gender identity, gender roles), and sexual orientation each uniquely shape the cognitive abilities reviewed. To date, however, few studies integrate these sex and gender factors together to better understand individual differences in cognitive functioning. This has potential benefits if a broader understanding of sex and gender factors are systematically measured when researching and treating numerous conditions where cognition is altered.
Collapse
Affiliation(s)
- Sarah Kheloui
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Silke Jacmin-Park
- Department of Psychology, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Ophélie Larocque
- Department of Psychology, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Philippe Kerr
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Mathias Rossi
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Louis Cartier
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada
| | - Robert-Paul Juster
- Department of Psychiatry and Addiction, University of Montreal, Montreal, Quebec, Canada; Department of Psychology, Université du Québec à Montréal, Montreal, Quebec, Canada; Centre de recherche de l'Institut universitaire en santé mentale de Montréal, Canada; Center on Sex⁎Gender, Allostasis and Resilience, Canada.
| |
Collapse
|
33
|
Lupu DC, Monedero I, Rodriguez-Ruiz C, Pita M, Turiegano E. In support of 2D:4D: More data exploring its conflicting results on handedness, sexual orientation and sex differences. PLoS One 2023; 18:e0280514. [PMID: 37607180 PMCID: PMC10443882 DOI: 10.1371/journal.pone.0280514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 12/28/2022] [Indexed: 08/24/2023] Open
Abstract
In the last few years, several studies have questioned the value of the second-to-fourth digit ratio (2D:4D) as a measure of exposure to sex hormones before birth. Controversy has also extended to the 2D:4D association with individual features previously related to this exposure such as handedness and sexual orientation. Given that it has been argued that sex differences in 2D:4D could be a consequence of body-size differences, we have tested in a large sample the allometric relationship between finger lengths and body size. Our results show that the association is either allometric or isometric, depending on the analyses performed. In any case, the deviation from isometry is not large enough to explain the typically observed sex difference in this trait. We have also tested the association between sexual orientation and 2D:4D, finding a relationship between 2D:4D and sexual orientation in men but not in women. We attribute this discordance with previously published meta-analysis to differences in genetic background, a variable that has gained relevance in recent years in studies involving 2D:4D. Finally, we did not find any relationship between 2D:4D and handedness, evaluated through self-reported preference and hand performance. Our main conclusion is that 2D:4D shows differences between sexes beyond their disparity in body size. In our opinion, 2D:4D can be used cautiously as an indicator of intrauterine exposure to sex hormones taking into account some considerations, such as analysing a very large sample and taking careful measurements of the ethnicity of the sample.
Collapse
Affiliation(s)
| | - Ignacio Monedero
- Departamento de Fisiología, Universidad Autónoma de Madrid, Madrid, España
| | | | - Miguel Pita
- Departamento de Biología, Universidad Autónoma de Madrid, Madrid, España
| | - Enrique Turiegano
- Departamento de Biología, Universidad Autónoma de Madrid, Madrid, España
| |
Collapse
|
34
|
Salia S, Martin Y, Burke FF, Myles LA, Jackman L, Halievski K, Bambico FR, Swift-Gallant A. Antibiotic-induced socio-sexual behavioral deficits are reversed via cecal microbiota transplantation but not androgen treatment. Brain Behav Immun Health 2023; 30:100637. [PMID: 37256194 PMCID: PMC10225889 DOI: 10.1016/j.bbih.2023.100637] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 04/03/2023] [Accepted: 05/07/2023] [Indexed: 06/01/2023] Open
Abstract
Recent evidence has demonstrated a sex-specific role of the gut microbiome on social behavior such as anxiety, possibly driven by a reciprocal relationship between the gut microbiome and gonadal hormones. For instance, gonadal hormones drive sex differences in gut microbiota composition, and certain gut bacteria can produce androgens from glucocorticoids. We thus asked whether the gut microbiome can influence androgen-dependent socio-sexual behaviors. We first treated C57BL/6 mice with broad-spectrum antibiotics (ABX) in drinking water to deplete the gut microbiota either transiently during early development (embryonic day 16-postnatal day [PND] 21) or in adulthood (PND 60-85). We hypothesized that if ABX interferes with androgens, then early ABX would interfere with critical periods for sexual differentiation of brain and thus lead to long-term decreases in males' socio-sexual behavior, while adult ABX would interfere with androgens' activational effects on behavior. We found that in males but not females, early and adult ABX treatment decreased territorial aggression, and adult ABX also decreased sexual odor preference. We then assessed whether testosterone and/or cecal microbiota transplantation (CMT) via oral gavage could prevent ABX-induced socio-sexual behavioral deficits in adult ABX-treated males. Mice were treated with same- or other-sex control cecum contents or with testosterone for two weeks. While testosterone was not effective in rescuing any behavior, we found that male CMT restored both olfactory preference and aggression in adult ABX male mice, while female CMT restored olfactory preference but not aggression. These results suggest sex-specific effects of the gut microbiome on socio-sexual behaviors, independent of androgens.
Collapse
|
35
|
Lozano Wun V, Foland‐Ross LC, Jo B, Green T, Hong D, Ross JL, Reiss AL. Adolescent brain development in girls with Turner syndrome. Hum Brain Mapp 2023; 44:4028-4039. [PMID: 37126641 PMCID: PMC10258525 DOI: 10.1002/hbm.26327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 02/08/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023] Open
Abstract
Turner syndrome (TS) is a common sex chromosome aneuploidy in females associated with various physical, cognitive, and socio-emotional phenotypes. However, few studies have examined TS-associated alterations in the development of cortical gray matter volume and the two components that comprise this measure-surface area and thickness. Moreover, the longitudinal direct (i.e., genetic) and indirect (i.e., hormonal) effects of X-monosomy on the brain are unclear. Brain structure was assessed in 61 girls with TS (11.3 ± 2.8 years) and 55 typically developing girls (10.8 ± 2.3 years) for up to 4 timepoints. Surface-based analyses of cortical gray matter volume, thickness, and surface area were conducted to examine the direct effects of X-monosomy present before pubertal onset and indirect hormonal effects of estrogen deficiency/X-monosomy emerging after pubertal onset. Longitudinal analyses revealed that, whereas typically developing girls exhibited normative declines in gray matter structure during adolescence, this pattern was reduced or inverted in TS. Further, girls with TS demonstrated smaller total surface area and larger average cortical thickness overall. Regionally, the TS group exhibited decreased volume and surface area in the pericalcarine, postcentral, and parietal regions relative to typically developing girls, as well as larger volume in the caudate, amygdala, and temporal lobe regions and increased thickness in parietal and temporal regions. Surface area alterations were predominant by age 8, while maturational differences in thickness emerged by age 10 or later. Taken together, these results suggest the involvement of both direct and indirect effects of X-chromosome haploinsufficiency on brain development in TS.
Collapse
Affiliation(s)
- Vanessa Lozano Wun
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
- Department of PsychologyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Lara C. Foland‐Ross
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
| | - Booil Jo
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
| | - Tamar Green
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
| | - David Hong
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
| | - Judith L. Ross
- Department of PediatricsThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Nemours Children's HospitalWilmingtonDelawareUSA
| | - Allan L. Reiss
- Center for Interdisciplinary Brain Sciences Research, Department of Psychiatry and Behavioral SciencesStanford UniversityStanfordCaliforniaUSA
- Department of PediatricsStanford University School of MedicineStanfordCaliforniaUSA
- Department of RadiologyStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
36
|
Le AL, Lynch WJ, Rissman EF. Sex Chromosome Complement and Estradiol Modify Cocaine Self-Administration Behaviors in Male Mice. Neuroendocrinology 2023; 113:1177-1188. [PMID: 37348474 DOI: 10.1159/000531648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 06/24/2023]
Abstract
INTRODUCTION Women are more vulnerable to cocaine's reinforcing effects and have a more rapid course to addiction after initial cocaine use as compared to men. Studies in rodents similarly indicate an enhanced sensitivity to the reinforcing effects of cocaine in females versus males. Levels of estradiol (E2) are correlated with vulnerability to the rewarding actions of cocaine. Here, we asked if sex chromosome complement (SCC) influences vulnerability to cocaine use. METHODS We used the four-core genotype mouse that produces gonadal males and females with either XX or XY SCC. Mice were gonadectomized and implanted with either an estradiol (E2) or cholesterol-filled pellet. This allowed us to determine the effects of SCC in the absence (cholesterol-treated) and presence of tonic high physiological hormone levels (estradiol). Acquisition of cocaine self-administration was determined over a 12-day period using an escalated dose procedure (0.3 mg/kg/infusion, sessions 1-6; 0.6 mg/kg/infusion, sessions 6-12). RESULTS Without estradiol treatment, a greater percentage of castrated XY mice acquired cocaine self-administration and did so at a faster rate than XX castrates and ovariectomized XY females. These same XY males acquired sooner, infused more cocaine, and directed more nose pokes to the rewarded nose-poke hole than XX castrates and XY males receiving E2. CONCLUSION Our results suggest that in gonadal male mice, SCC and estradiol can modulate the reinforcing effects of cocaine which may influence the likelihood of cocaine use.
Collapse
Affiliation(s)
- Aaron L Le
- Department of Biological Sciences, Center for Human Health and the Environment, NCSU, Raleigh, North Carolina, USA
| | - Wendy J Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Emilie F Rissman
- Department of Biological Sciences, Center for Human Health and the Environment, NCSU, Raleigh, North Carolina, USA
| |
Collapse
|
37
|
Zhao Z, Gobrogge K. Neurodevelopmental Model Explaining Associations between Sex Hormones, Personality, and Eating Pathology. Brain Sci 2023; 13:859. [PMID: 37371339 PMCID: PMC10296733 DOI: 10.3390/brainsci13060859] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Clinical scientists have been investigating the relationships between sex hormones, personality, and eating disorders for decades. However, there is a lack of direct research that addresses whether personality mediates or moderates the relationships between sex hormones and eating pathology. Moreover, the neural mechanisms that underlie the interactive associations between these variables remain unclear. This review aims to summarize the associations between these constructs, describe a neural mechanism mediating these relationships, and offer clinical strategies for the early identification and intervention of eating disorders. The gathered evidence shows that aggressiveness, impulsivity, and obsessive-compulsiveness may mediate or moderate the relationships between sex hormones and eating pathology, but only among females. Furthermore, sex hormone receptor density in the mesocorticolimbic dopamine pathway may explain the neural mechanism of these associations. Future research should use more comprehensive personality measurements and assess the mediation and moderation effects of temperament while taking the hormone levels of women across menstrual cycles into account. Additionally, electroencephalography and functional magnetic resonance imaging should be implemented to directly assess brain activity and corroborate these findings.
Collapse
Affiliation(s)
- Ziyu Zhao
- Department of Psychological & Brain Sciences, Boston University, Boston, MA 02215, USA;
| | - Kyle Gobrogge
- Department of Psychological & Brain Sciences, Boston University, Boston, MA 02215, USA;
- Undergraduate Program in Neuroscience, College of Art & Sciences, Boston University, Boston, MA 02215, USA
| |
Collapse
|
38
|
Jing X, Lyu L, Gong Y, Wen H, Li Y, Wang X, Li J, Yao Y, Zuo C, Xie S, Yan S, Qi X. Olfactory receptor OR52N2 for PGE 2 in mediation of guppy courtship behaviors. Int J Biol Macromol 2023; 241:124518. [PMID: 37088189 DOI: 10.1016/j.ijbiomac.2023.124518] [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: 06/15/2022] [Revised: 01/19/2023] [Accepted: 04/15/2023] [Indexed: 04/25/2023]
Abstract
Prostaglandins (PGs) are a type of physiologically active unsaturated fatty acids. As an important sex pheromone, PGs play a vital role in regulating the reproductive behaviors of species by mediating nerve and endocrine responses. In this study, guppy (Poecilia reticulate) was used as the model specie to detect the function of PGE2 in inducing the onset of courtship behaviors. Our results showed that adding PGE2 into the water environment could activate the courtship behavior of male guppy, indicating that the peripheral olfactory system mediated the PGE2 function. Thereafter, the open reading frame (ORF) of olfactory receptor or52n2 was cloned, which was 936 bp in length, coding 311 amino acids. As a typical G protein-coupled receptor, OR52N2 had a conservative seven α-helix transmembrane domains. To confirm the regulatory relationship between OR52N2 and PGE2, dual-luciferase reporter assay was employed to verify the activation of downstream CREB signaling pathways. Results showed that PGE2 significantly enhanced CRE promoter activity in or52n2 ORF transient transfected HEK-293 T cells. Finally, localization of or52n2 mRNA were observed in ciliated receptor cells of the olfactory epithelium using in situ hybridization. Our results provide a novel insight into sex pheromone signaling transduction in reproductive behavior.
Collapse
Affiliation(s)
- Xiao Jing
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Likang Lyu
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yu Gong
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Haishen Wen
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yun Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xiaojie Wang
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Jianshuang Li
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yijia Yao
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Chenpeng Zuo
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Songyang Xie
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Shaojing Yan
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xin Qi
- Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China.
| |
Collapse
|
39
|
Conlon FL, Arnold AP. Sex chromosome mechanisms in cardiac development and disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:340-350. [PMID: 37808586 PMCID: PMC10558115 DOI: 10.1038/s44161-023-00256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 02/13/2023] [Indexed: 10/10/2023]
Abstract
Many human diseases, including cardiovascular disease, show differences between men and women in pathology and treatment outcomes. In the case of cardiac disease, sex differences are exemplified by differences in the frequency of specific types of congenital and adult-onset heart disease. Clinical studies have suggested that gonadal hormones are a factor in sex bias. However, recent research has shown that gene and protein networks under non-hormonal control also account for cardiac sex differences. In this review, we describe the sex chromosome pathways that lead to sex differences in the development and function of the heart and highlight how these findings affect future care and treatment of cardiac disease.
Collapse
Affiliation(s)
- Frank L Conlon
- Departments of Biology and Genetics, McAllister Heart Institute, UNC-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Arthur P Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
40
|
Lischinsky JE, Yin L, Shi C, Prakash N, Burke J, Shekaran G, Grba M, Corbin JG, Lin D. Hardwired to attack: Transcriptionally defined amygdala subpopulations play distinct roles in innate social behaviors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532692. [PMID: 36993508 PMCID: PMC10055059 DOI: 10.1101/2023.03.16.532692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Social behaviors are innate and supported by dedicated neural circuits, but it remains unclear whether these circuits are developmentally hardwired or established through social experience. Here, we revealed distinct response patterns and functions in social behavior of medial amygdala (MeA) cells originating from two embryonically parcellated developmental lineages. MeA cells in male mice that express the transcription factor Foxp2 (MeAFoxp2) are specialized for processing male conspecific cues even before puberty and are essential for adult inter-male aggression. In contrast, MeA cells derived from the Dbx1-lineage (MeADbx1) respond broadly to social cues and are non-essential for male aggression. Furthermore, MeAFoxp2 and MeADbx1 cells show differential anatomical and functional connectivity. Altogether, our results support a developmentally hardwired aggression circuit at the level of the MeA and we propose a lineage-based circuit organization by which a cell's embryonic transcription factor profile determines its social information representation and behavior relevance during adulthood.
Collapse
Affiliation(s)
- Julieta E Lischinsky
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Luping Yin
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Chenxi Shi
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Hunter College, New York, NY, USA
| | - Nandkishore Prakash
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, United States
| | - Jared Burke
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Govind Shekaran
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Maria Grba
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, United States
| | - Dayu Lin
- Neuroscience Institute, New York University School of Medicine, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
41
|
Colón L, Peru E, Zuloaga DG, Poulos AM. Contributions of gonadal hormones in the sex-specific organization of context fear learning. PLoS One 2023; 18:e0282293. [PMID: 36862730 PMCID: PMC9980802 DOI: 10.1371/journal.pone.0282293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/11/2023] [Indexed: 03/03/2023] Open
Abstract
It is widely established that gonadal hormones are fundamental to modulating and organizing the sex-specific nature of reproductive behaviors. Recently we proposed that context fear conditioning (CFC) may emerge in a sex-specific manner organized prior to the pubertal surge of gonadal hormones. Here we sought to determine the necessity of male and female gonadal hormones secreted at critical periods of development upon context fear learning. We tested the organizational hypothesis that neonatal and pubertal gonadal hormones play a permanent role in organizing contextual fear learning. We demonstrate that the postnatal absence of gonadal hormones by neonatal orchiectomy (oRX) in males and ovariectomy (oVX) in females resulted in an attenuation of CFC in adult males and an enhancement of CFC in adult females. In females, the gradual introduction of estrogen before conditioning partially rescued this effect. However, the decrease of CFC in adult males was not rescued by introducing testosterone before conditioning. Next, at a further point in development, preventing the pubertal surge of gonadal hormones by prepubertal oRX in males resulted in a reduction in adult CFC. In contrast, in females, prepubertal oVX did not alter adult CFC. However, the adult introduction of estrogen in prepubertal oVX rats reduced adult CFC. Lastly, the adult-specific deletion of gonadal hormones by adult oRX or oVX alone or replacement of testosterone or estrogen did not alter CFC. Consistent with our hypothesis, we provide initial evidence that gonadal hormones at early periods of development exert a vital role in the organization and development of CFC in male and female rats.
Collapse
Affiliation(s)
- Lorianna Colón
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, United States of America
| | - Eduardo Peru
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, United States of America
| | - Damian G. Zuloaga
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, United States of America
| | - Andrew M. Poulos
- Department of Psychology and Center for Neuroscience Research, State University of New York at Albany, Albany, NY, United States of America
- * E-mail:
| |
Collapse
|
42
|
Mulvey B, Frye HE, Lintz T, Fass S, Tycksen E, Nelson EC, Morón JA, Dougherty JD. Cnih3 Deletion Dysregulates Dorsal Hippocampal Transcription across the Estrous Cycle. eNeuro 2023; 10:ENEURO.0153-22.2023. [PMID: 36849260 PMCID: PMC10027183 DOI: 10.1523/eneuro.0153-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023] Open
Abstract
In females, the hippocampus, a critical brain region for coordination of learning, memory, and behavior, displays altered physiology and behavioral output across the estrous or menstrual cycle. However, the molecular effectors and cell types underlying these observed cyclic changes have only been partially characterized to date. Recently, profiling of mice null for the AMPA receptor trafficking gene Cnih3 have demonstrated estrous-dependent phenotypes in dorsal hippocampal synaptic plasticity, composition, and learning/memory. We therefore profiled dorsal hippocampal transcriptomes from female mice in each estrous cycle stage, and contrasted it with that of males, across wild-type (WT) and Cnih3 mutants. In wild types, we identified only subtle differences in gene expression between the sexes, while comparing estrous stages to one another revealed up to >1000 differentially expressed genes (DEGs). These estrous-responsive genes are especially enriched in gene markers of oligodendrocytes and the dentate gyrus, and in functional gene sets relating to estrogen response, potassium channels, and synaptic gene splicing. Surprisingly, Cnih3 knock-outs (KOs) showed far broader transcriptomic differences between estrous cycle stages and males. Moreover, Cnih3 knock-out drove subtle but extensive expression changes accentuating sex differential expression at diestrus and estrus. Altogether, our profiling highlights cell types and molecular systems potentially impacted by estrous-specific gene expression patterns in the adult dorsal hippocampus, enabling mechanistic hypothesis generation for future studies of sex-differential neuropsychiatric function and dysfunction. Moreover, these findings suggest an unrecognized role of Cnih3 in buffering against transcriptional effects of estrous, providing a candidate molecular mechanism to explain estrous-dependent phenotypes observed with Cnih3 loss.
Collapse
Affiliation(s)
- Bernard Mulvey
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Hannah E Frye
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tania Lintz
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Stuart Fass
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Eric Tycksen
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| | - Jose A Morón
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
43
|
Neufeld SAS, Collaer ML, Spencer D, Pasterski V, Hindmarsh PC, Hughes IA, Acerini C, Hines M. Androgens and child behavior: Color and toy preferences in children with congenital adrenal hyperplasia (CAH). Horm Behav 2023; 149:105310. [PMID: 36738514 PMCID: PMC9976950 DOI: 10.1016/j.yhbeh.2023.105310] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023]
Abstract
Human males and females show average gender/sex differences for certain psychological phenomena. Multiple factors may contribute to these differences, including sex chromosomes, exposure to gonadal hormones, and socialization or learning. This study investigated potential hormonal and socialization/learning influences on gender/sex differences in childhood preferences for color, specifically pink and red vs. blues, and for toys. Children (aged 4 to 11 years) with congenital adrenal hyperplasia (CAH, n = 43 girls and 37 boys), marked by elevated prenatal adrenal androgen exposure, and without CAH (n = 41 girls and 31 boys) were studied. Prior research indicates girls with CAH are masculinized for certain behaviors, such as toy choices, while boys with CAH generally do not differ from boys without CAH. In the current study, children indicated preferences for stereotyped hues of pink vs. blue as well as two control color pairs. They also indicated their preference between gender/sex-typed toys (doll vs. car) presented in black and white, in gender/sex-congruent colors (pink doll vs. blue car) and in gender/sex-incongruent colors (pink car vs. blue doll). Color findings: Control girls preferred stereotyped pink over blue more than boys or girls with CAH did; the latter two groups did not differ in their color preferences. No preference differences occurred for other color pairs. Toy findings: In black/white or gender/sex-congruent colors, boys preferred the car more than control girls or girls with CAH did, while girls with CAH preferred the car more than control girls did. In gender/sex-incongruent colors (pink car vs. blue doll), boys still preferred the car, while girls with CAH showed reduced and control girls showed increased preferences for the pink car compared to the car preferences in black/white. Results support learning theories of color preferences, perhaps also influenced by pre-existing toy preferences which may occur for other reasons, including early androgen exposure. Specifically, girls with CAH may have learned they do not enjoy stereotypical toys for girls, often colored pink, and pink coloring may subsequently diminish their preference for a car. Our results highlight the importance of gonadal hormones and learning in the development of childhood toy and color preferences.
Collapse
Affiliation(s)
- Sharon A S Neufeld
- Department of Psychology, University of Cambridge, Downing Place, Cambridge CB2 3EB, UK.
| | - Marcia L Collaer
- Department of Psychology and Neuroscience Program, Middlebury College, Middlebury, VT 05753, USA.
| | - Debra Spencer
- Department of Psychology, University of Cambridge, Downing Place, Cambridge CB2 3EB, UK.
| | - Vickie Pasterski
- Department of Psychology, University of Cambridge, Downing Place, Cambridge CB2 3EB, UK; Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Peter C Hindmarsh
- Developmental Endocrinology Research Group, Institute of Child Health, University College London, London, UK.
| | - Ieuan A Hughes
- Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
| | - Carlo Acerini
- Department of Paediatrics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | - Melissa Hines
- Department of Psychology, University of Cambridge, Downing Place, Cambridge CB2 3EB, UK.
| |
Collapse
|
44
|
Hashimoto JG, Singer ML, Goeke CM, Zhang F, Song Y, Xia K, Linhardt RJ, Guizzetti M. Sex differences in hippocampal structural plasticity and glycosaminoglycan disaccharide levels after neonatal handling. Exp Neurol 2023; 361:114313. [PMID: 36572372 PMCID: PMC10097408 DOI: 10.1016/j.expneurol.2022.114313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
In this study we investigated the effects of a neonatal handling protocol that mimics the handling of sham control pups in protocols of neonatal exposure to brain insults on dendritic arborization and glycosaminoglycan (GAG) levels in the developing brain. GAGs are long, unbranched polysaccharides, consisting of repeating disaccharide units that can be modified by sulfation at specific sites and are involved in modulating neuronal plasticity during brain development. In this study, male and female Sprague-Dawley rats underwent neonatal handling daily between post-natal day (PD)4 and PD9, with brains analyzed on PD9. Neuronal morphology and morphometric analysis of the apical and basal dendritic trees of CA1 hippocampal pyramidal neurons were carried out by Golgi-Cox staining followed by neuron tracing and analysis with the software Neurolucida. Chondroitin sulfate (CS)-, Hyaluronic Acid (HA)-, and Heparan Sulfate (HS)-GAG disaccharide levels were quantified in the hippocampus by Liquid Chromatography/Mass Spectrometry analyses. We found sex by neonatal handling interactions on several parameters of CA1 pyramidal neuron morphology and in the levels of HS-GAGs, with females, but not males, showing an increase in both dendritic arborization and HS-GAG levels. We also observed increased expression of glucocorticoid receptor gene Nr3c1 in the hippocampus of both males and females following neonatal handling suggesting that both sexes experienced a similar stress during the handling procedure. This is the first study to show sex differences in two parameters of brain plasticity, CA1 neuron morphology and HS-GAG levels, following handling stress in neonatal rats.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Mo L Singer
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Calla M Goeke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Yuefan Song
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Ke Xia
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
45
|
Arnold AP, Chen X, Grzybowski MN, Ryan JM, Sengelaub DR, Mohanroy T, Furlan VA, Grisham W, Malloy L, Takizawa A, Wiese CB, Vergnes L, Skaletsky H, Page DC, Reue K, Harley VR, Dwinell MR, Geurts AM. A "Four Core Genotypes" rat model to distinguish mechanisms underlying sex-biased phenotypes and diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527738. [PMID: 36798326 PMCID: PMC9934672 DOI: 10.1101/2023.02.09.527738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Background We have generated a rat model similar to the Four Core Genotypes mouse model, allowing comparison of XX and XY rats with the same type of gonad. The model detects novel sex chromosome effects (XX vs. XY) that contribute to sex differences in any rat phenotype. Methods XY rats were produced with an autosomal transgene of Sry , the testis-determining factor gene, which were fathers of XX and XY progeny with testes. In other rats, CRISPR-Cas9 technology was used to remove Y chromosome factors that initiate testis differentiation, producing fertile XY gonadal females that have XX and XY progeny with ovaries. These groups can be compared to detect sex differences caused by sex chromosome complement (XX vs. XY) and/or by gonadal hormones (rats with testes vs. ovaries). Results We have measured numerous phenotypes to characterize this model, including gonadal histology, breeding performance, anogenital distance, levels of reproductive hormones, body and organ weights, and central nervous system sexual dimorphisms. Serum testosterone levels were comparable in adult XX and XY gonadal males. Numerous phenotypes previously found to be sexually differentiated by the action of gonadal hormones were found to be similar in XX and XY rats with the same type of gonad, suggesting that XX and XY rats with the same type of gonad have comparable levels of gonadal hormones at various stages of development. Conclusion The results establish a powerful new model to discriminate sex chromosome and gonadal hormone effects that cause sexual differences in rat physiology and disease.
Collapse
|
46
|
Valencia-Olvera AC, Maldonado Weng J, Christensen A, LaDu MJ, Pike CJ. Role of estrogen in women's Alzheimer's disease risk as modified by APOE. J Neuroendocrinol 2023; 35:e13209. [PMID: 36420620 PMCID: PMC10049970 DOI: 10.1111/jne.13209] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is characterized by numerous sexual dimorphisms that impact the development, progression, and probably the strategies to prevent and treat the most common form of dementia. In this review, we consider this topic from a female perspective with a specific focus on how women's vulnerability to the disease is affected by the individual and interactive effects of estrogens and apolipoprotein E (APOE) genotype. Importantly, APOE appears to modulate systemic and neural outcomes of both menopause and estrogen-based hormone therapy. In the brain, dementia risk is greater in APOE4 carriers, and the impacts of hormone therapy on cognitive decline and dementia risk vary according to both outcome measure and APOE genotype. Beyond the CNS, estrogen and APOE genotype affect vulnerability to menopause-associated bone loss, dyslipidemia and cardiovascular disease risk. An emerging concept that may link these relationships is the possibility that the effects of APOE in women interact with estrogen status by mechanisms that may include modulation of estrogen responsiveness. This review highlights the need to consider the key AD risk factors of advancing age in a sex-specific manner to optimize development of therapeutic approaches for AD, a view aligned with the principle of personalized medicine.
Collapse
Affiliation(s)
- AC Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - J Maldonado Weng
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - A Christensen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| | - MJ LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - CJ Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089 USA
| |
Collapse
|
47
|
Krüger THC, Leeners B, Tronci E, Mancini T, Ille F, Egli M, Engler H, Röblitz S, Frieling H, Sinke C, Jahn K. The androgen system across the menstrual cycle: Hormonal, (epi-)genetic and psychometric alterations. Physiol Behav 2023; 259:114034. [PMID: 36403781 DOI: 10.1016/j.physbeh.2022.114034] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/09/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022]
Abstract
The menstrual cycle is characterized by various hormonal alterations and associations with mental and physical conditions have been postulated. Among endocrine factors, the androgen system has been a target of major interest in males and to a lesser extent in females and may influence emotion, cognition, behavior and somatic factors. Only few studies investigated alterations of these parameters throughout the menstrual cycle and there is a lack of studies exploring a link towards epigenetic and genetic regulation. This multisite longitudinal study examines behavioral parameters including affectivity, stress perception and various diary parameters of mental and physical well-being in conjunction with testosterone and LH plasma levels in 87 menstruating women. Additionally, Cysteine-Adenenine-Guanin (CAG) repeat length and methylation of the androgen receptor gene collected at four time points across two cycles comprising the menstrual, pre-ovulatory, mid-luteal and premenstrual phase were assesed. There was a significant increase of LH and testosterone plasma levels during the pre-ovulatory phase as well as a decrease of methylation of the androgen receptor at mid-luteal phase. Subjective ratings of physical condition and sexual interest peaked during the pre-ovulatory phase and the former correlated negatively with the androgen receptor gene methylation level. This longitudinal study shows alterations of the androgen system including epigenetic measurements throughout the menstrual cycle. While a link between peripheral testosterone and sexual activity and between increased physical condition and an upregulation of testosterone receptor protein expression can be assumed, the majority of parameters remained unchanged. These initial findings need validation by subsequent studies.
Collapse
Affiliation(s)
- Tillmann H C Krüger
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Section of Clinical Psychology and Sexual Medicine, Medical School Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Brigitte Leeners
- Department of Reproductive Endocrinology, University Hospital Zürich, Switzerland
| | - Enrico Tronci
- Department of Computer Science, Sapienza University of Rome, Italy
| | - Toni Mancini
- Department of Computer Science, Sapienza University of Rome, Italy
| | - Fabian Ille
- Center of Competence in Aerospace Biomedical Science & Technology, Lucerne University of Applied Sciences and Arts, Switzerland
| | - Marcel Egli
- Center of Competence in Aerospace Biomedical Science & Technology, Lucerne University of Applied Sciences and Arts, Switzerland
| | - Harald Engler
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, Germany
| | - Susanna Röblitz
- Computational Biology Unit, Department of Informatics,University of Bergen, Norway
| | - Helge Frieling
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Laboratory of Molecular Psychiatry, Hannover Medical School, Hannover, Germany
| | - Christopher Sinke
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Section of Clinical Psychology and Sexual Medicine, Medical School Hannover, Germany
| | - Kirsten Jahn
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Laboratory of Molecular Psychiatry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
48
|
Hampson E. Oral contraceptives in the central nervous system: Basic pharmacology, methodological considerations, and current state of the field. Front Neuroendocrinol 2023; 68:101040. [PMID: 36243109 DOI: 10.1016/j.yfrne.2022.101040] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/19/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022]
Abstract
Millions of women around the world use combined oral contraceptives (OCs), yet surprisingly little is known about their central nervous system (CNS) effects. This article provides a short overview of the basic pharmacology of OCs, emphasizing features that may be relevant to understanding their effects in the CNS. Historical and recent findings from studies of cognitive function, mood, and negative affect (depressive changes under OC use) are then reviewed. We also present data from an archival dataset from our own laboratory in which we explore dysphoric changes in women using four generations of contraceptive progestins. Current data in the field are consistent with a modest effect of OC use on CNS variables, but conclusions based on current findings must be made very cautiously because of multiple methodological issues in many published studies to date, and inconsistencies in the findings. Directions for future research over the next 10 years are suggested. (150 words).
Collapse
Affiliation(s)
- Elizabeth Hampson
- Department of Psychology, University of Western Ontario, London, ON, Canada; Department of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
49
|
Edwards PD, Toor I, Holmes MM. The Curious Case of the Naked Mole-Rat: How Extreme Social and Reproductive Adaptations Might Influence Sex Differences in the Brain. Curr Top Behav Neurosci 2023; 62:47-70. [PMID: 35301704 DOI: 10.1007/7854_2022_310] [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: 10/18/2022]
Abstract
Research in the neurobiology of sex differences is inherently influenced by the study species that are used. Some traditional animal research models, such as rats and mice, show certain sex differences in the brain that have been foundational to neurobiological research. However, subsequent work has demonstrated that these differences are not always generalizable, especially to species with different social structures and sex-associated roles or behaviors. One such example is the naked mole-rat (Heterocephalus glaber), which has an unusual social structure among mammals. Naked mole-rats live in large groups where reproduction is restricted to a dominant female, called the "queen," and often only one breeding male. All other animals in the group, the "subordinates," are socially suppressed from reproduction and remain in a prepubescent state as adults, unless they are removed from the presence of the queen. These subordinates show little to no sex differences in external morphology, neural morphology, or behavior. However, there are a suite of neurobiological differences between subordinate and breeding naked mole-rats. After naked mole-rats attain breeding status, many of the classically sexually differentiated brain regions increase in volume (paraventricular nucleus, medial amygdala, bed nucleus of the stria terminalis). There are additionally social status differences in sex hormone receptor expression in the brain, as well as other changes in gene expression, some of which also show sex differences - though not always in the predicted direction based on other rodent studies. Data from naked mole-rats show that it is critical to consider the evolved social structure of a species when studying sex differences in the brain.
Collapse
Affiliation(s)
- Phoebe D Edwards
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Ilapreet Toor
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Melissa M Holmes
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada.
| |
Collapse
|
50
|
Mikhail ME, Carroll SL, Clark DA, O’Connor SM, Culbert KM, Burt SA, Klump KL. Disadvantage and disordered eating in boys: Examining phenotypic and genotype × environment associations across development. JOURNAL OF PSYCHOPATHOLOGY AND CLINICAL SCIENCE 2023; 132:51-62. [PMID: 36689370 PMCID: PMC9878376 DOI: 10.1037/abn0000791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Socioeconomic disadvantage may be a significant risk factor for disordered eating, particularly for individuals with underlying genetic risk. However, little to nothing is known about the impact of disadvantage on disordered eating in boys during the critical developmental risk period. Crucially, risk models developed for girls may not necessarily apply to boys, as boys show different developmental patterns of disordered eating risk (i.e., earlier activation of genetic influences during adrenarche, an early stage of puberty). This is the first study to examine phenotypic and Genotype × Environment (G × E) effects of disadvantage in boys. Analyses examined 3,484 male twins ages 8-17 (Mage = 12.27, SD = 2.96) from the Michigan State University Twin Registry. Disordered eating (e.g., body dissatisfaction, binge eating) was measured using the parent-report Michigan Twins Project Eating Disorder Survey. Neighborhood disadvantage was measured using a census-tract level Area Deprivation Index, and family socioeconomic status was determined from parental income and education. Adrenarche status was determined using multiple indicators, including age and Pubertal Development Scale scores. G × E models suggested that genetic influences on disordered eating were activated earlier for boys experiencing familial or neighborhood disadvantage, with substantial genetic influences in early adrenarche, when genetic influences were low in more advantaged boys. Phenotypically, both neighborhood and familial disadvantage were associated with greater disordered eating for boys in late adrenarche, which could indicate a lasting impact of earlier activation of genetic influences on later risk. Results highlight disadvantage as a novel risk factor for disordered eating in boys, particularly those with genetic vulnerabilities. (PsycInfo Database Record (c) 2023 APA, all rights reserved).
Collapse
Affiliation(s)
- Megan E. Mikhail
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Sarah L. Carroll
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - D. Angus Clark
- Department of Psychiatry and Addiction Center, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Kristen M. Culbert
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - S. Alexandra Burt
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Kelly L. Klump
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|