1
|
Henechowicz TL, Coleman PL, Gustavson DE, Mekki YN, Nayak S, Nitin R, Scartozzi AC, Tio ES, van Klei R, Felsky D, Thaut MH, Gordon RL. Polygenic Associations between Motor Behaviour, Neuromotor Traits, and Active Music Engagement in Four Cohorts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645667. [PMID: 40196524 PMCID: PMC11974849 DOI: 10.1101/2025.03.27.645667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Phenotypic investigations have shown that actively engaging with music, i.e., playing a musical instrument or singing may be protective of motor decline in aging. For example, music training associated with enhanced sensorimotor skills accompanied by changes in brain structure and function. Although it is possible that the benefits of active music engagement "transfer" to benefits in the motor domain, it is also possible that the genetic architecture of motor behaviour and the motor system structure may influence active music engagement. This study investigated whether polygenic scores (PGS) for five behavioural motor traits, 12 neuromotor structural brain traits, and seven rates of change in brain structure traits trained from existing discovery genome-wide association studies (GWAS) predict active music engagement outcomes in four independent cohorts of unrelated individuals of European ancestry: the Canadian Longitudinal Study on Aging (CLSA; N=22,198), Wisconsin Longitudinal Study (WLS; N=4,605), Vanderbilt's BioVU Repository (BioVU; N=6,150), and Vanderbilt's Online Musicality study (OM; N=1,559). Results were meta-analyzed for each PGS main effect across outcomes and cohorts, revealing that PGS for a faster walking pace was associated with higher amounts of active music engagement. Within CLSA, a higher PGS for walking pace was associated with greater odds of engaging with music. Findings suggest a shared genetic architecture between motor function and active music engagement. Future intervention-based research should consider the genetic underpinnings of motor behavior when evaluating the effects of music engagement on motor function across the lifespan.
Collapse
Affiliation(s)
- T L Henechowicz
- Music and Health Science Research Collaboratory, Faculty of Music, University of Toronto
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
- Music Cognition Laboratory, Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center
| | - P L Coleman
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
- Center for Digital Genomic Medicine, Vanderbilt University Medical Center
| | - D E Gustavson
- Institute for Behavioral Genetics, University of Colorado Boulder
| | - Y N Mekki
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
| | - S Nayak
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
- Music Cognition Laboratory, Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center
| | - R Nitin
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
| | - A C Scartozzi
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
- Music Cognition Laboratory, Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center
| | - E S Tio
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health
| | - R van Klei
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health
| | - D Felsky
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health
- Department of Psychiatry, University of Toronto
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto
- Rotman Research Institute, Baycrest Hospital, Toronto, ON
- Department of Anthropology, University of Toronto
| | - M H Thaut
- Music and Health Science Research Collaboratory, Faculty of Music, University of Toronto
- Temerty Faculty of Medicine, University of Toronto
| | - R L Gordon
- Music and Health Science Research Collaboratory, Faculty of Music, University of Toronto
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center
- Music Cognition Laboratory, Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center
- Center for Digital Genomic Medicine, Vanderbilt University Medical Center
- Institute for Behavioral Genetics, University of Colorado Boulder
- Department of Psychiatry, University of Toronto
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto
- Rotman Research Institute, Baycrest Hospital, Toronto, ON
- Department of Anthropology, University of Toronto
- Temerty Faculty of Medicine, University of Toronto
- Vanderbilt Brain Institute, Vanderbilt University
- Department of Psychology, Vanderbilt University
| |
Collapse
|
2
|
Snell DM, Turner JMA. X chromosome passed from mother to daughter influences brain ageing. Nature 2025; 638:45-47. [PMID: 39843690 DOI: 10.1038/d41586-025-00079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
|
3
|
Jiang Z, Sullivan PF, Li T, Zhao B, Wang X, Luo T, Huang S, Guan PY, Chen J, Yang Y, Stein JL, Li Y, Liu D, Sun L, Zhu H. The X chromosome's influences on the human brain. SCIENCE ADVANCES 2025; 11:eadq5360. [PMID: 39854466 PMCID: PMC11759047 DOI: 10.1126/sciadv.adq5360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025]
Abstract
Genes on the X chromosome are extensively expressed in the human brain. However, little is known for the X chromosome's impact on the brain anatomy, microstructure, and functional networks. We examined 1045 complex brain imaging traits from 38,529 participants in the UK Biobank. We unveiled potential autosome-X chromosome interactions while proposing an atlas outlining dosage compensation for brain imaging traits. Through extensive association studies, we identified 72 genome-wide significant trait-locus pairs (including 29 new associations) that share genetic architectures with brain-related disorders, notably schizophrenia. Furthermore, we found unique sex-specific associations and assessed variations in genetic effects between sexes. Our research offers critical insights into the X chromosome's role in the human brain, underscoring its contribution to the differences observed in brain structure and functionality between sexes.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick F. Sullivan
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tengfei Li
- Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xifeng Wang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tianyou Luo
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Shuai Huang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Y. Guan
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jie Chen
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yue Yang
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jason L. Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dajiang Liu
- Department of Public Health Sciences, Penn State University, Hershey, PA 17033, USA
- Department of Biochemistry and Molecular Biology, Penn State University, Hershey, PA 17033, USA
| | - Lei Sun
- Department of Statistical Sciences, University of Toronto, Toronto, ON M5G 1Z5, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Biomedical Research Imaging Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Statistics and Operations Research, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
4
|
Gui Y, Zhou G, Cui S, Li H, Lu H, Zhao H. The left amygdala is genetically sexually-dimorphic: multi-omics analysis of structural MRI volumes. Transl Psychiatry 2025; 15:17. [PMID: 39843917 PMCID: PMC11754786 DOI: 10.1038/s41398-025-03223-8] [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: 03/19/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Brain anatomy plays a key role in complex behaviors and mental disorders that are sexually divergent. While our understanding of the sex differences in the brain anatomy remains relatively limited, particularly of the underlying genetic and molecular mechanisms that contribute to these differences. We performed the largest study of sex differences in brain volumes (N = 33,208) by examining sex differences both in the raw brain volumes and after controlling the whole brain volumes. Genetic correlation analysis revealed sex differences only in the left amygdala. We compared transcriptome differences between males and females using data from GTEx and characterized cell-type compositions using GTEx bulk amygdala RNA-seq data and LIBD amygdala single-cell reference profiles. We also constructed polygenic risk scores (PRS) to investigate sex-specific genetic correlations between left amygdala volume and mental disorders (N = 25,576~105,318) of Psychiatric Genomics Consortium and other traits of UKB (N = 347,996). Although there were pronounced sex differences in brain volumes, there was no difference in the heritability between sexes. There was a significant sex-specific genetic correlation between male and female left amygdala. We identified sex-differentiated genetic effects of PRSs for schizophrenia on left amygdala volume, as well as significant sex-differentiated genetic correlations between PRSs of left amygdala and six traits in UKB. We also found several sex-differentially expressed genes in the amygdala. These findings not only advanced the current knowledge of genetic basis of sex differences in brain anatomy, but also presented an important clue for future research on the mechanism of sex differences in mental disorders and targeted treatments.
Collapse
Affiliation(s)
- Yuanyuan Gui
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Geyu Zhou
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Shuya Cui
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyu Li
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Hui Lu
- SJTU-Yale Joint Center for Biostatistics and Data Science, Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Hongyu Zhao
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
5
|
Chaumeil J, Morey C. [X chromosome regulation and female functional specificities: Are two Xs better than one?]. Med Sci (Paris) 2024; 40:935-946. [PMID: 39705564 DOI: 10.1051/medsci/2024179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
What if the presence of two X chromosomes confers functional specificities on female cells and contributes to the different susceptibilites of men and women to certain diseases? One of the X chromosomes is randomly silenced in each female cell from the embryonic stage, theoretically making the sexes equal. This silencing of the X chromosome is a unique epigenetic process, affecting an entire chromosome and resulting in mosaic expression of X-linked genes throughout the body. However, some genes escape this process and X-inactivation appears to be somewhat labile in certain cell types. What are the physiological implications of these observations? This question is beginning to be explored, particularly in the immune and nervous systems, where several pathologies have sexual bias.
Collapse
Affiliation(s)
- Julie Chaumeil
- Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris, France
| | - Céline Morey
- UMR7216 - Épigénétique et destin cellulaire, CNRS, Université Paris Cité, Paris, France
| |
Collapse
|
6
|
Serrarens C, Kashyap S, Otter M, Campforts BCM, Stumpel CTRM, Linden DEJ, van Amelsvoort TAMJ, Vingerhoets C. White matter organization abnormalities in adults with 47,XXX: A 7 Tesla MRI study. Psychiatry Res Neuroimaging 2024; 345:111915. [PMID: 39546963 DOI: 10.1016/j.pscychresns.2024.111915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
47,XXX (Triple X syndrome) is a sex chromosome aneuploidy characterized by the presence of a supernumerary X chromosome in affected females, and has been associated with a variable cognitive, behavioral, and psychiatric phenotype. Alterations in brain gray matter structure and function have been reported, but less is known about white matter (WM) organization in 47,XXX. Therefore, we conducted 7 T diffusion tensor imaging and characterized fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity of 22 adult women with 47,XXX and 22 age-matched typically developing females using tract-based spatial statistics. Relationships between phenotypic traits and WM organization characteristics in 47,XXX were also investigated. Adults with 47,XXX showed lower axial diffusivity in the body of the corpus callosum and the right superior longitudinal fasciculus. WM organization variability was not associated with IQ and social cognition and social functioning deficits in 47,XXX. Our findings indicate an effect of a supernumerary X chromosome in adult women on axonal integrity of the body of the corpus callosum and the right superior longitudinal fasciculus. These findings provide additional insight into the role of the X chromosome on WM organization. Future research is warranted to explore the clinical significant impact of altered WM organization in 47,XXX.
Collapse
Affiliation(s)
- Chaira Serrarens
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands.
| | - Sriranga Kashyap
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands; Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Maarten Otter
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands; Medical Department, SIZA, Arnhem, the Netherlands
| | - Bea C M Campforts
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Constance T R M Stumpel
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - David E J Linden
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Thérèse A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands
| | - Claudia Vingerhoets
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, the Netherlands; 's Heeren Loo Zorggroep, Amersfoort, the Netherlands
| |
Collapse
|
7
|
Gozlan E, Lewit-Cohen Y, Frenkel D. Sex Differences in Astrocyte Activity. Cells 2024; 13:1724. [PMID: 39451242 PMCID: PMC11506538 DOI: 10.3390/cells13201724] [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: 08/15/2024] [Revised: 09/27/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Astrocytes are essential for maintaining brain homeostasis. Alterations in their activity have been associated with various brain pathologies. Sex differences were reported to affect astrocyte development and activity, and even susceptibility to different neurodegenerative diseases. This review aims to summarize the current knowledge on the effects of sex on astrocyte activity in health and disease.
Collapse
Affiliation(s)
- Elisa Gozlan
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
| | - Yarden Lewit-Cohen
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
| | - Dan Frenkel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.G.); (Y.L.-C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
8
|
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
|
9
|
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, TorontoONM5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, TorontoONM5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
| | - Taylor DeYoung
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, TorontoONM5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
| | - A. Elizabeth de Guzman
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, TorontoONM5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, RoveretoTN38068, Italy
| | - Madeline W. Elder
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
| | - Antoine Beauchamp
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
| | - C. Shun Wong
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Odette Cancer Centre, TorontoONM4N 3M5, Canada
- Department of Radiation Oncology, University of Toronto, TorontoONM5T 1P5, Canada
| | - Mark R. Palmert
- Division of Endocrinology, The Hospital for Sick Children, University of Toronto, TorontoONM5G 1X8, Canada
- Department of Pediatrics, University of Toronto, TorontoONM5S 1A8, Canada
- Department of Physiology, University of Toronto, TorontoONM5S 1A8, Canada
- Genetics and Genome Biology, Hospital for Sick Children, TorontoONM5G 1X8, Canada
| | - Jason P. Lerch
- Mouse Imaging Centre, Hospital for Sick Children, TorontoONM5T 3H7, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
- Wellcome Centre for Integrative Neuroimaging, Medical Sciences Division, University of Oxford, Oxford, OXFOX3 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, TorontoONM5T 3H7, Canada
- Translational Medicine, Hospital for Sick Children, TorontoONM5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, TorontoONM5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ONM5G 0A3, Canada
| |
Collapse
|
10
|
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. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608129. [PMID: 39229144 PMCID: PMC11370326 DOI: 10.1101/2024.08.15.608129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
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
Affiliation(s)
- Carinna Torgerson
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Katherine Bottenhorn
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Hedyeh Ahmadi
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jeiran Choupan
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
- NeuroScope Inc., New York, USA
| | - Megan M. Herting
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
11
|
Serrarens C, Ruiz-Fernandez J, Otter M, Campforts BCM, Stumpel CTRM, Linden DEJ, van Amelsvoort TAMJ, Kashyap S, Vingerhoets C. Intracortical myelin across laminae in adult individuals with 47,XXX: a 7 Tesla MRI study. Cereb Cortex 2024; 34:bhae343. [PMID: 39183364 PMCID: PMC11345119 DOI: 10.1093/cercor/bhae343] [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: 04/12/2024] [Revised: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024] Open
Abstract
47,XXX (Triple X syndrome) is a sex chromosome aneuploidy characterized by the presence of a supernumerary X chromosome in affected females and is associated with a variable cognitive, behavioral, and psychiatric phenotype. The effect of a supernumerary X chromosome in affected females on intracortical microstructure is currently unknown. Therefore, we conducted 7 Tesla structural MRI and compared T1 (ms), as a proxy for intracortical myelin (ICM), across laminae of 21 adult women with 47,XXX and 22 age-matched typically developing females using laminar analyses. Relationships between phenotypic traits and T1 values in 47,XXX were also investigated. Adults with 47,XXX showed higher bilateral T1 across supragranular laminae in the banks of the superior temporal sulcus, and in the right inferior temporal gyrus, suggesting decreases of ICM primarily within the temporal cortex in 47,XXX. Higher social functioning in 47,XXX was related to larger inferior temporal gyrus ICM content. Our findings indicate an effect of a supernumerary X chromosome in adult-aged women on ICM across supragranular laminae within the temporal cortex. These findings provide insight into the role of X chromosome dosage on ICM across laminae. Future research is warranted to further explore the functional significance of altered ICM across laminae in 47,XXX.
Collapse
Affiliation(s)
- Chaira Serrarens
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Julia Ruiz-Fernandez
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
- INSERM U1299, Centre Borelli UMR 9010, ENS-Paris-Saclay, Université Paris Saclay, Paris, France
| | - Maarten Otter
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
- Medical Department, SIZA, Arnhem, 6800 AM, The Netherlands
| | - Bea C M Campforts
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Constance T R M Stumpel
- Department of Clinical Genetics and School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, 6229 ER, The Netherlands
| | - David E J Linden
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Therese A M J van Amelsvoort
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Sriranga Kashyap
- Department of Cognitive Neuroscience, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, 6229 EV, The Netherlands
- Krembil Brain Institute, University Health Network, Toronto, ON M5T 2S8, Canada
| | - Claudia Vingerhoets
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Institute (MHeNS), Maastricht University, Maastricht, 6200 MD, The Netherlands
- ‘s Heeren Loo Zorggroep, Amersfoort, 3818 LA, The Netherlands
| |
Collapse
|
12
|
Montani C, Balasco L, Pagani M, Alvino FG, Barsotti N, de Guzman AE, Galbusera A, de Felice A, Nickl-Jockschat TK, Migliarini S, Casarosa S, Lau P, Mattioni L, Pasqualetti M, Provenzano G, Bozzi Y, Lombardo MV, Gozzi A. Sex-biasing influence of autism-associated Ube3a gene overdosage at connectomic, behavioral, and transcriptomic levels. SCIENCE ADVANCES 2024; 10:eadg1421. [PMID: 38996019 PMCID: PMC11244557 DOI: 10.1126/sciadv.adg1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/07/2024] [Indexed: 07/14/2024]
Abstract
Genomic mechanisms enhancing risk in males may contribute to sex bias in autism. The ubiquitin protein ligase E3A gene (Ube3a) affects cellular homeostasis via control of protein turnover and by acting as transcriptional coactivator with steroid hormone receptors. Overdosage of Ube3a via duplication or triplication of chromosomal region 15q11-13 causes 1 to 2% of autistic cases. Here, we test the hypothesis that increased dosage of Ube3a may influence autism-relevant phenotypes in a sex-biased manner. We show that mice with extra copies of Ube3a exhibit sex-biasing effects on brain connectomics and autism-relevant behaviors. These effects are associated with transcriptional dysregulation of autism-associated genes, as well as genes differentially expressed in 15q duplication and in autistic people. Increased Ube3a dosage also affects expression of genes on the X chromosome, genes influenced by sex steroid hormone, and genes sex-differentially regulated by transcription factors. These results suggest that Ube3a overdosage can contribute to sex bias in neurodevelopmental conditions via influence on sex-differential mechanisms.
Collapse
Affiliation(s)
- Caterina Montani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Luigi Balasco
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
- IMT School for Advanced Studies, Lucca, Italy
| | - Filomena Grazia Alvino
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Noemi Barsotti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - A. Elizabeth de Guzman
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alessia de Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Thomas K. Nickl-Jockschat
- Department of Psychiatry and Psychotherapy, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Mental Health (DZPG), partner site Halle-Jena-Magdeburg, Germany
- Center for Intervention and Research on adaptive and maladaptive brain Circuits underlying mental health (C-I-R-C), Halle-Jena-Magdeburg, Germany
| | - Sara Migliarini
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Simona Casarosa
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Centre for Medical Sciences (CISMed), University of Trento, Trento, Italy
| | - Pierre Lau
- Istituto Italiano di Tecnologia, Center for Human Technologies, Genova, Italy
| | - Lorenzo Mattioni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Massimo Pasqualetti
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Rovereto, Italy
- CNR Neuroscience Institute, Pisa, Italy
| | - Michael V. Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, CNCS@UNITN, Rovereto, Italy
| |
Collapse
|
13
|
Burmistrov DE, Gudkov SV, Franceschi C, Vedunova MV. Sex as a Determinant of Age-Related Changes in the Brain. Int J Mol Sci 2024; 25:7122. [PMID: 39000227 PMCID: PMC11241365 DOI: 10.3390/ijms25137122] [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] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The notion of notable anatomical, biochemical, and behavioral distinctions within male and female brains has been a contentious topic of interest within the scientific community over several decades. Advancements in neuroimaging and molecular biological techniques have increasingly elucidated common mechanisms characterizing brain aging while also revealing disparities between sexes in these processes. Variations in cognitive functions; susceptibility to and progression of neurodegenerative conditions, notably Alzheimer's and Parkinson's diseases; and notable disparities in life expectancy between sexes, underscore the significance of evaluating aging within the framework of gender differences. This comprehensive review surveys contemporary literature on the restructuring of brain structures and fundamental processes unfolding in the aging brain at cellular and molecular levels, with a focus on gender distinctions. Additionally, the review delves into age-related cognitive alterations, exploring factors influencing the acceleration or deceleration of aging, with particular attention to estrogen's hormonal support of the central nervous system.
Collapse
Affiliation(s)
- Dmitriy E. Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilova St., 119991 Moscow, Russia;
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| | - Maria V. Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603022 Nizhny Novgorod, Russia
| |
Collapse
|
14
|
Jiang Z, Sullivan PF, Li T, Zhao B, Wang X, Luo T, Huang S, Guan PY, Chen J, Yang Y, Stein JL, Li Y, Liu D, Sun L, Zhu H. The pivotal role of the X-chromosome in the genetic architecture of the human brain. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.30.23294848. [PMID: 37693466 PMCID: PMC10491353 DOI: 10.1101/2023.08.30.23294848] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Genes on the X-chromosome are extensively expressed in the human brain. However, little is known for the X-chromosome's impact on the brain anatomy, microstructure, and functional network. We examined 1,045 complex brain imaging traits from 38,529 participants in the UK Biobank. We unveiled potential autosome-X-chromosome interactions, while proposing an atlas outlining dosage compensation (DC) for brain imaging traits. Through extensive association studies, we identified 72 genome-wide significant trait-locus pairs (including 29 new associations) that share genetic architectures with brain-related disorders, notably schizophrenia. Furthermore, we discovered unique sex-specific associations and assessed variations in genetic effects between sexes. Our research offers critical insights into the X-chromosome's role in the human brain, underscoring its contribution to the differences observed in brain structure and functionality between sexes.
Collapse
|
15
|
Pottmeier P, Nikolantonaki D, Lanner F, Peuckert C, Jazin E. Sex-biased gene expression during neural differentiation of human embryonic stem cells. Front Cell Dev Biol 2024; 12:1341373. [PMID: 38764741 PMCID: PMC11101176 DOI: 10.3389/fcell.2024.1341373] [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: 11/20/2023] [Accepted: 04/16/2024] [Indexed: 05/21/2024] Open
Abstract
Sex differences in the developing human brain are primarily attributed to hormonal influence. Recently however, genetic differences and their impact on the developing nervous system have attracted increased attention. To understand genetically driven sexual dimorphisms in neurodevelopment, we investigated genome-wide gene expression in an in vitro differentiation model of male and female human embryonic stem cell lines (hESC), independent of the effects of human sex hormones. Four male and four female-derived hESC lines were differentiated into a population of mixed neurons over 37 days. Differential gene expression and gene set enrichment analyses were conducted on bulk RNA sequencing data. While similar differentiation tendencies in all cell lines demonstrated the robustness and reproducibility of our differentiation protocol, we found sex-biased gene expression already in undifferentiated ESCs at day 0, but most profoundly after 37 days of differentiation. Male and female cell lines exhibited sex-biased expression of genes involved in neurodevelopment, suggesting that sex influences the differentiation trajectory. Interestingly, the highest contribution to sex differences was found to arise from the male transcriptome, involving both Y chromosome and autosomal genes. We propose 13 sex-biased candidate genes (10 upregulated in male cell lines and 3 in female lines) that are likely to affect neuronal development. Additionally, we confirmed gene dosage compensation of X/Y homologs escaping X chromosome inactivation through their Y homologs and identified a significant overexpression of the Y-linked demethylase UTY and KDM5D in male hESC during neuron development, confirming previous results in neural stem cells. Our results suggest that genetic sex differences affect neuronal differentiation trajectories, which could ultimately contribute to sex biases during human brain development.
Collapse
Affiliation(s)
- Philipp Pottmeier
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Danai Nikolantonaki
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Fredrik Lanner
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Christiane Peuckert
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Elena Jazin
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| |
Collapse
|
16
|
Torgerson C, Ahmadi H, Choupan J, Fan CC, Blosnich JR, Herting MM. Sex, gender diversity, and brain structure in early adolescence. Hum Brain Mapp 2024; 45:e26671. [PMID: 38590252 PMCID: PMC11002534 DOI: 10.1002/hbm.26671] [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: 07/28/2023] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
There remains little consensus about the relationship between sex and brain structure, particularly in early adolescence. Moreover, few pediatric neuroimaging studies have analyzed both sex and gender as variables of interest-many of which included small sample sizes and relied on binary definitions of gender. The current study examined gender diversity with a continuous felt-gender score and categorized sex based on X and Y allele frequency in a large sample of children ages 9-11 years old (N = 7195). Then, a statistical model-building approach was employed to determine whether gender diversity and sex independently or jointly relate to brain morphology, including subcortical volume, cortical thickness, gyrification, and white matter microstructure. Additional sensitivity analyses found that male versus female differences in gyrification and white matter were largely accounted for by total brain volume, rather than sex per se. The model with sex, but not gender diversity, was the best-fitting model in 60.1% of gray matter regions and 61.9% of white matter regions after adjusting for brain volume. The proportion of variance accounted for by sex was negligible to small in all cases. While models including felt-gender explained a greater amount of variance in a few regions, the felt-gender score alone was not a significant predictor on its own for any white or gray matter regions examined. Overall, these findings demonstrate that at ages 9-11 years old, sex accounts for a small proportion of variance in brain structure, while gender diversity is not directly associated with neurostructural diversity.
Collapse
Affiliation(s)
- Carinna Torgerson
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Mark and Mary Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hedyeh Ahmadi
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jeiran Choupan
- Mark and Mary Stevens Neuroimaging and Informatics InstituteUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Chun Chieh Fan
- Center for Population Neuroscience and GeneticsLaureate Institute for Brain ResearchTulsaOklahomaUSA
- Department of Radiology, School of MedicineUniversity of CaliforniaSan DiegoCaliforniaUSA
| | - John R. Blosnich
- Suzanne Dworak‐Peck School of Social WorkUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Megan M. Herting
- Department of Population and Public Health SciencesUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
17
|
Siqueiros-Sanchez M, Rai B, Chowdhury S, Reiss AL, Green T. Syndrome-Specific Neuroanatomical Phenotypes in Girls With Turner and Noonan Syndromes. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2024; 9:146-155. [PMID: 36084900 PMCID: PMC10305746 DOI: 10.1016/j.bpsc.2022.08.012] [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: 04/11/2022] [Revised: 07/20/2022] [Accepted: 08/25/2022] [Indexed: 06/03/2023]
Abstract
BACKGROUND Turner syndrome (TS) and Noonan syndrome (NS) are distinct genetic conditions with highly similar physical and neurodevelopmental phenotypes. TS is caused by X chromosome absence, whereas NS results from genetic mutations activating the Ras-mitogen-activated protein kinase signaling pathway. Previous neuroimaging studies in individuals with TS and NS have shown neuroanatomical variations relative to typically developing individuals, a standard comparison group when initially examining a clinical group of interest. However, none of these studies included a second clinical comparison group, limiting their ability to identify syndrome-specific neuroanatomical phenotypes. METHODS In this study, we compared the behavioral and brain phenotypes of 37 girls with TS, 26 girls with NS, and 37 typically developing girls, all ages 5 to 12 years, using univariate and multivariate data-driven analyses. RESULTS We found divergent neuroanatomical phenotypes between groups, despite high behavioral similarities. Relative to the typically developing group, TS was associated with smaller whole-brain cortical surface area (p ≤ .0001), whereas NS was associated with smaller whole-brain cortical thickness (p = .013). TS was associated with larger subcortical volumes (left amygdala, p = .002; right hippocampus, p = .002), whereas NS was associated with smaller subcortical volumes (bilateral caudate, p ≤ .003; putamen, p < .001; pallidum, p < .001; right hippocampus, p = .015). Multivariate analyses also showed diverging brain phenotypes in terms of surface area and cortical thickness, with surface area outperforming cortical thickness at group separation. CONCLUSIONS TS and NS have syndrome-specific brain phenotypes, despite their behavioral similarities. Our observations suggest that neuroanatomical phenotypes better reflect the different genetic etiologies of TS and NS and may be superior biomarkers relative to behavioral phenotypes.
Collapse
Affiliation(s)
- Monica Siqueiros-Sanchez
- Brain Imaging, Development and Genetics Lab, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Bhavana Rai
- Brain Imaging, Development and Genetics Lab, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Samir Chowdhury
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Brain Dynamics Lab, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California
| | - Allan L Reiss
- Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Department of Radiology, Stanford University School of Medicine, Stanford University, Stanford, California; Department Pediatrics, Stanford University School of Medicine, Stanford University, Stanford, California
| | - Tamar Green
- Brain Imaging, Development and Genetics Lab, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California; Division of Interdisciplinary Brain Sciences, Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
18
|
Hanson C, Blumenthal J, Clasen L, Guma E, Raznahan A. Influences of sex chromosome aneuploidy on height, weight, and body mass index in human childhood and adolescence. Am J Med Genet A 2024; 194:150-159. [PMID: 37768018 DOI: 10.1002/ajmg.a.63398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/21/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023]
Abstract
Sex chromosome aneuploidies (SCAs) are collectively common conditions caused by carriage of a sex chromosome dosage other than XX for females and XY for males. Increases in sex chromosome dosage (SCD) have been shown to have an inverted-U association with height, but we lack combined studies of SCA effects on height and weight, and it is not known if any such effects vary with age. Here, we study norm-derived height and weight z-scores in 177 youth spanning 8 SCA karyotypes (XXX, XXY, XYY, XXXX, XXXY, XXYY, XXXXX, and XXXXY). We replicate a previously described inverted-U association between mounting SCD and height, and further show that there is also a muted version of this effect for weight: both phenotypes are elevated until SCD reaches 4 for females and 5 for males but decrease thereafter. We next use 266 longitudinal measures available from a subset of karyotypes (XXX, XXY, XYY, and XXYY) to show that mean height in these SCAs diverges further from norms with increasing age. As weight does not diverge from norms with increasing age, BMI decreases with increasing age. These findings extend our understanding of growth as an important clinical outcome in SCA, and as a key context for known effects of SCA on diverse organ systems that scale with body size.
Collapse
Affiliation(s)
- Claire Hanson
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland, USA
| | - Jonathan Blumenthal
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland, USA
| | - Liv Clasen
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland, USA
| | - Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland, USA
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health Intramural Research Program, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Farrell K, Auerbach A, Liu C, Martin K, Pareno M, Ray WK, Helm RF, Biase F, Jarome TJ. Sex-differences in proteasome-dependent K48-polyubiquitin signaling in the amygdala are developmentally regulated in rats. Biol Sex Differ 2023; 14:80. [PMID: 37950270 PMCID: PMC10638793 DOI: 10.1186/s13293-023-00566-z] [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: 06/28/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Sex differences have been observed in several brain regions for the molecular mechanisms involved in baseline (resting) and memory-related processes. The ubiquitin proteasome system (UPS) is a major protein degradation pathway in cells. Sex differences have been observed in lysine-48 (K48)-polyubiquitination, the canonical degradation mark of the UPS, both at baseline and during fear memory formation within the amygdala. Here, we investigated when, how, and why these baseline sex differences arise and whether both sexes require the K48-polyubiquitin mark for memory formation in the amygdala. METHODS We used a combination of molecular, biochemical and proteomic approaches to examine global and protein-specific K48-polyubiquitination and DNA methylation levels at a major ubiquitin coding gene (Uba52) at baseline in the amygdala of male and female rats before and after puberty to determine if sex differences were developmentally regulated. We then used behavioral and genetic approaches to test the necessity of K48-polyubiquitination in the amygdala for fear memory formation. RESULTS We observed developmentally regulated baseline differences in Uba52 methylation and total K48-polyubiquitination, with sexual maturity altering levels specifically in female rats. K48-polyubiquitination at specific proteins changed across development in both male and female rats, but sex differences were present regardless of age. Lastly, we found that genetic inhibition of K48-polyubiquitination in the amygdala of female, but not male, rats impaired fear memory formation. CONCLUSIONS These results suggest that K48-polyubiquitination differentially targets proteins in the amygdala in a sex-specific manner regardless of age. However, sexual maturity is important in the developmental regulation of K48-polyubiquitination levels in female rats. Consistent with these data, K48-polyubiquitin signaling in the amygdala is selectively required to form fear memories in female rats. Together, these data indicate that sex-differences in baseline K48-polyubiquitination within the amygdala are developmentally regulated, which could have important implications for better understanding sex-differences in molecular mechanisms involved in processes relevant to anxiety-related disorders such as post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Kayla Farrell
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA
| | - Aubrey Auerbach
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Catherine Liu
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA
| | - Kiley Martin
- School of Neuroscience, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA
| | - Myasia Pareno
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA
| | - W Keith Ray
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Richard F Helm
- Department of Biochemistry, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Fernando Biase
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA
| | - Timothy J Jarome
- School of Animal Sciences, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA.
- School of Neuroscience, Virginia Polytechnic Institute and State University, 175 West Campus Dr., 2150 Litton-Reaves Hall, Blacksburg, VA, 24061, USA.
| |
Collapse
|
20
|
VanderLaan DP, Skorska MN, Peragine DE, Coome LA. Carving the Biodevelopment of Same-Sex Sexual Orientation at Its Joints. ARCHIVES OF SEXUAL BEHAVIOR 2023; 52:2939-2962. [PMID: 35960401 DOI: 10.1007/s10508-022-02360-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 06/15/2023]
Abstract
Sexual orientation is a core aspect of human experience and understanding its development is fundamental to psychology as a scientific discipline. Biological perspectives have played an important role in uncovering the processes that contribute to sexual orientation development. Research in this field has relied on a variety of populations, including community, clinical, and cross-cultural samples, and has commonly focused on female gynephilia (i.e., female sexual attraction to adult females) and male androphilia (i.e., male sexual attraction to adult males). Genetic, hormonal, and immunological processes all appear to influence sexual orientation. Consistent with biological perspectives, there are sexual orientation differences in brain development and evidence indicates that similar biological influences apply across cultures. An outstanding question in the field is whether the hypothesized biological influences are all part of the same process or represent different developmental pathways leading to same-sex sexual orientation. Some studies indicate that same-sex sexually oriented people can be divided into subgroups who likely experienced different biological influences. Consideration of gender expression in addition to sexual orientation might help delineate such subgroups. Thus, future research on the possible existence of such subgroups could prove to be valuable for uncovering the biological development of sexual orientation. Recommendations for such future research are discussed.
Collapse
Affiliation(s)
- Doug P VanderLaan
- Department of Psychology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON, L5L 1C6, Canada.
- Child and Youth Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada.
| | - Malvina N Skorska
- Child and Youth Psychiatry, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Diana E Peragine
- Department of Psychology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON, L5L 1C6, Canada
| | - Lindsay A Coome
- Department of Psychology, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON, L5L 1C6, Canada
| |
Collapse
|
21
|
Torgerson C, Ahmadi H, Choupan J, Fan CC, Blosnich JR, Herting MM. Sex, gender diversity, and brain structure in children ages 9 to 11 years old. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551036. [PMID: 37546960 PMCID: PMC10402171 DOI: 10.1101/2023.07.28.551036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
There remains little consensus about the relationship between sex and brain structure, particularly in childhood. Moreover, few pediatric neuroimaging studies have analyzed both sex and gender as variables of interest - many of which included small sample sizes and relied on binary definitions of gender. The current study examined gender diversity with a continuous felt-gender score and categorized sex based on X and Y allele frequency in a large sample of children ages 9-11 years-old (N=7693). Then, a statistical model-building approach was employed to determine whether gender diversity and sex independently or jointly relate to brain morphology, including subcortical volume, cortical thickness, gyrification, and white matter microstructure. The model with sex, but not gender diversity, was the best-fitting model in 75% of gray matter regions and 79% of white matter regions examined. The addition of gender to the sex model explained significantly more variance than sex alone with regard to bilateral cerebellum volume, left precentral cortical thickness, as well as gyrification in the right superior frontal gyrus, right parahippocampal gyrus, and several regions in the left parietal lobe. For mean diffusivity in the left uncinate fasciculus, the model with sex, gender, and their interaction captured the most variance. Nonetheless, the magnitude of variance accounted for by sex was small in all cases and felt-gender score was not a significant predictor on its own for any white or gray matter regions examined. Overall, these findings demonstrate that at ages 9-11 years-old, sex accounts for a small proportion of variance in brain structure, while gender diversity is not directly associated with neurostructural diversity.
Collapse
Affiliation(s)
- Carinna Torgerson
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Hedyeh Ahmadi
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| | - Jeiran Choupan
- Mark and Mary Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, USA
| | - Chun Chieh Fan
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, OK, USA
- Department of Radiology, School of Medicine, University of California, San Diego
| | - John R. Blosnich
- Suzanne Dworak-Peck School of Social Work, University of Southern California, Los Angeles, CA, USA
| | - Megan M. Herting
- Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
22
|
Raheel K, Deegan G, Di Giulio I, Cash D, Ilic K, Gnoni V, Chaudhuri KR, Drakatos P, Moran R, Rosenzweig I. Sex differences in alpha-synucleinopathies: a systematic review. Front Neurol 2023; 14:1204104. [PMID: 37545736 PMCID: PMC10398394 DOI: 10.3389/fneur.2023.1204104] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/13/2023] [Indexed: 08/08/2023] Open
Abstract
Background Past research indicates a higher prevalence, incidence, and severe clinical manifestations of alpha-synucleinopathies in men, leading to a suggestion of neuroprotective properties of female sex hormones (especially estrogen). The potential pathomechanisms of any such effect on alpha-synucleinopathies, however, are far from understood. With that aim, we undertook to systematically review, and to critically assess, contemporary evidence on sex and gender differences in alpha-synucleinopathies using a bench-to-bedside approach. Methods In this systematic review, studies investigating sex and gender differences in alpha-synucleinopathies (Rapid Eye Movement (REM) Behavior Disorder (RBD), Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA)) from 2012 to 2022 were identified using electronic database searches of PubMed, Embase and Ovid. Results One hundred sixty-two studies were included; 5 RBD, 6 MSA, 20 DLB and 131 PD studies. Overall, there is conclusive evidence to suggest sex-and gender-specific manifestation in demographics, biomarkers, genetics, clinical features, interventions, and quality of life in alpha-synucleinopathies. Only limited data exists on the effects of distinct sex hormones, with majority of studies concentrating on estrogen and its speculated neuroprotective effects. Conclusion Future studies disentangling the underlying sex-specific mechanisms of alpha-synucleinopathies are urgently needed in order to enable novel sex-specific therapeutics.
Collapse
Affiliation(s)
- Kausar Raheel
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Gemma Deegan
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
| | - Irene Di Giulio
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
| | - Diana Cash
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Katarina Ilic
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- BRAIN, Imaging Centre, CNS, King’s College London, London, United Kingdom
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Valentina Gnoni
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Center for Neurodegenerative Diseases and the Aging Brain, University of Bari Aldo Moro, Lecce, Italy
| | - K. Ray Chaudhuri
- Movement Disorders Unit, King’s College Hospital and Department of Clinical and Basic Neurosciences, Institute of Psychiatry, Psychology and Neuroscience and Parkinson Foundation Centre of Excellence, King’s College London, London, United Kingdom
| | - Panagis Drakatos
- School of Basic and Medical Biosciences, Faculty of Life Science and Medicine, King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Rosalyn Moran
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, United Kingdom
- Sleep Disorders Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
23
|
Yang F, Chen WZ, Jiang SS, Wang XH, Xu RS. A candidate protective factor in amyotrophic lateral sclerosis: heterogenous nuclear ribonucleoprotein G. Neural Regen Res 2023; 18:1527-1534. [PMID: 36571358 PMCID: PMC10075103 DOI: 10.4103/1673-5374.357916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Heterogenous nuclear ribonucleoprotein G is down-regulated in the spinal cord of the Tg(SOD1*G93A)1Gur (TG) amyotrophic lateral sclerosis mouse model. However, most studies have only examined heterogenous nuclear ribonucleoprotein G expression in the amyotrophic lateral sclerosis model and heterogenous nuclear ribonucleoprotein G effects in amyotrophic lateral sclerosis pathogenesis such as in apoptosis are unknown. In this study, we studied the potential mechanism of heterogenous nuclear ribonucleoprotein G in neuronal death in the spinal cord of TG and wild-type mice and examined the mechanism by which heterogenous nuclear ribonucleoprotein G induces apoptosis. Heterogenous nuclear ribonucleoprotein G in spinal cord was analyzed using immunohistochemistry and western blotting, and cell proliferation and proteins (TAR DNA binding protein 43, superoxide dismutase 1, and Bax) were detected by the Cell Counting Kit-8 and western blot analysis in heterogenous nuclear ribonucleoprotein G siRNA-transfected PC12 cells. We analyzed heterogenous nuclear ribonucleoprotein G distribution in spinal cord in the amyotrophic lateral sclerosis model at various time points and the expressions of apoptosis and proliferation-related proteins. Heterogenous nuclear ribonucleoprotein G was mainly localized in neurons. Amyotrophic lateral sclerosis mice were examined at three stages: preonset (60-70 days), onset (90-100 days) and progression (120-130 days). The number of heterogenous nuclear ribonucleoprotein G-positive cells was significantly higher in the anterior horn of the lumbar spinal cord segment of TG mice at the preonset stage than that of control group but lower than that of the control group at the onset stage. The number of heterogenous nuclear ribonucleoprotein G-positive cells in both central canal and surrounding gray matter of the whole spinal cord of TG mice at the onset stage was significantly lower than that in the control group, whereas that of the lumbar spinal cord segment of TG mice was significantly higher than that in the control group at preonset stage and significantly lower than that in the control group at the progression stage. The numbers of heterogenous nuclear ribonucleoprotein G-positive cells in the posterior horn of cervical and thoracic segments of TG mice at preonset and progression stages were significantly lower than those in the control group. The expression of heterogenous nuclear ribonucleoprotein G in the cervical spinal cord segment of TG mice was significantly higher than that in the control group at the preonset stage but significantly lower at the progression stage. The expression of heterogenous nuclear ribonucleoprotein G in the thoracic spinal cord segment of TG mice was significantly increased at the preonset stage, significantly decreased at the onset stage, and significantly increased at the progression stage compared with the control group. heterogenous nuclear ribonucleoprotein G expression in the lumbar spinal cord segment of TG mice was significantly lower than that of the control group at the progression stage. After heterogenous nuclear ribonucleoprotein G gene silencing, PC12 cell survival was lower than that of control cells. Both TAR DNA binding protein 43 and Bax expressions were significantly increased in heterogenous nuclear ribonucleoprotein G-silenced cells compared with control cells. Our study suggests that abnormal distribution and expression of heterogenous nuclear ribonucleoprotein G might play a protective effect in amyotrophic lateral sclerosis development via preventing neuronal death by reducing abnormal TAR DNA binding protein 43 generation in the spinal cord.
Collapse
Affiliation(s)
- Fang Yang
- Department of Neurology, Jiangxi Provincial People's Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Wen-Zhi Chen
- Department of Neurology, Jiangxi Provincial People's Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Shi-Shi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Xiao-Hua Wang
- Department of Geriatrics and General Practice/General Family Medicine, Jiangxi Provincial People's Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, China
| | - Ren-Shi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, China
| |
Collapse
|
24
|
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 PMCID: PMC10704933 DOI: 10.1159/000531648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [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
- Center for Human Health and the Environment, Department of Biological Sciences, NCSU, Raleigh, NC 27695
| | - Wendy J. Lynch
- Department of Psychiatry and Neurobehavioral Sciences, University of Virginia School of Medicine, Charlottesville VA 22903
| | - Emilie F. Rissman
- Center for Human Health and the Environment, Department of Biological Sciences, NCSU, Raleigh, NC 27695
| |
Collapse
|
25
|
Calvo N, Einstein G. Steroid hormones: risk and resilience in women's Alzheimer disease. Front Aging Neurosci 2023; 15:1159435. [PMID: 37396653 PMCID: PMC10313425 DOI: 10.3389/fnagi.2023.1159435] [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: 02/05/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
More women have Alzheimer disease (AD) than men, but the reasons for this phenomenon are still unknown. Including women in clinical research and studying their biology is key to understand not just their increased risk but also their resilience against the disease. In this sense, women are more affected by AD than men, but their reserve or resilience mechanisms might delay symptom onset. The aim of this review was to explore what is known about mechanisms underlying women's risk and resilience in AD and identify emerging themes in this area that merit further research. We conducted a review of studies analyzing molecular mechanisms that may induce neuroplasticity in women, as well as cognitive and brain reserve. We also analyzed how the loss of steroid hormones in aging may be linked to AD. We included empirical studies with human and animal models, literature reviews as well as meta-analyses. Our search identified the importance of 17-b-estradiol (E2) as a mechanism driving cognitive and brain reserve in women. More broadly, our analysis revealed the following emerging perspectives: (1) the importance of steroid hormones and their effects on both neurons and glia for the study of risk and resilience in AD, (2) E2's crucial role in women's brain reserve, (3) women's verbal memory advantage as a cognitive reserve factor, and (4) E2's potential role in linguistic experiences such as multilingualism and hearing loss. Future directions for research include analyzing the reserve mechanisms of steroid hormones on neuronal and glial plasticity, as well as identifying the links between steroid hormone loss in aging and risk for AD.
Collapse
Affiliation(s)
- Noelia Calvo
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Gillian Einstein
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Tema Genus, Linköping University, Linköping, Sweden
- Women’s College Research Institute, Toronto, ON, Canada
- Centre for Life Course and Aging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Qian SH, Xiong YL, Chen L, Geng YJ, Tang XM, Chen ZX. Dynamic Spatial-temporal Expression Ratio of X Chromosome to Autosomes but Stable Dosage Compensation in Mammals. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:589-600. [PMID: 36031057 PMCID: PMC10787176 DOI: 10.1016/j.gpb.2022.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 08/03/2022] [Accepted: 08/22/2022] [Indexed: 06/15/2023]
Abstract
In the evolutionary model of dosage compensation, per-allele expression level of the X chromosome has been proposed to have twofold up-regulation to compensate its dose reduction in males (XY) compared to females (XX). However, the expression regulation of X-linked genes is still controversial, and comprehensive evaluations are still lacking. By integrating multi-omics datasets in mammals, we investigated the expression ratios including X to autosomes (X:AA ratio) and X to orthologs (X:XX ratio) at the transcriptome, translatome, and proteome levels. We revealed a dynamic spatial-temporal X:AA ratio during development in humans and mice. Meanwhile, by tracing the evolution of orthologous gene expression in chickens, platypuses, and opossums, we found a stable expression ratio of X-linked genes in humans to their autosomal orthologs in other species (X:XX ≈ 1) across tissues and developmental stages, demonstrating stable dosage compensation in mammals. We also found that different epigenetic regulations contributed to the high tissue specificity and stage specificity of X-linked gene expression, thus affecting X:AA ratios. It could be concluded that the dynamics of X:AA ratios were attributed to the different gene contents and expression preferences of the X chromosome, rather than the stable dosage compensation.
Collapse
Affiliation(s)
- Sheng Hu Qian
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yu-Li Xiong
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lu Chen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Ying-Jie Geng
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao-Man Tang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen-Xia Chen
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China; Interdisciplinary Sciences Institute, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
27
|
Terra Machado D, Bernardes Brustolini OJ, Côrtes Martins Y, Grivet Mattoso Maia MA, Ribeiro de Vasconcelos AT. Inference of differentially expressed genes using generalized linear mixed models in a pairwise fashion. PeerJ 2023; 11:e15145. [PMID: 37033732 PMCID: PMC10078460 DOI: 10.7717/peerj.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Background
Technological advances involving RNA-Seq and Bioinformatics allow quantifying the transcriptional levels of genes in cells, tissues, and cell lines, permitting the identification of Differentially Expressed Genes (DEGs). DESeq2 and edgeR are well-established computational tools used for this purpose and they are based upon generalized linear models (GLMs) that consider only fixed effects in modeling. However, the inclusion of random effects reduces the risk of missing potential DEGs that may be essential in the context of the biological phenomenon under investigation. The generalized linear mixed models (GLMM) can be used to include both effects.
Methods
We present DEGRE (Differentially Expressed Genes with Random Effects), a user-friendly tool capable of inferring DEGs where fixed and random effects on individuals are considered in the experimental design of RNA-Seq research. DEGRE preprocesses the raw matrices before fitting GLMMs on the genes and the derived regression coefficients are analyzed using the Wald statistical test. DEGRE offers the Benjamini-Hochberg or Bonferroni techniques for P-value adjustment.
Results
The datasets used for DEGRE assessment were simulated with known identification of DEGs. These have fixed effects, and the random effects were estimated and inserted to measure the impact of experimental designs with high biological variability. For DEGs’ inference, preprocessing effectively prepares the data and retains overdispersed genes. The biological coefficient of variation is inferred from the counting matrices to assess variability before and after the preprocessing. The DEGRE is computationally validated through its performance by the simulation of counting matrices, which have biological variability related to fixed and random effects. DEGRE also provides improved assessment measures for detecting DEGs in cases with higher biological variability. We show that the preprocessing established here effectively removes technical variation from those matrices. This tool also detects new potential candidate DEGs in the transcriptome data of patients with bipolar disorder, presenting a promising tool to detect more relevant genes.
Conclusions
DEGRE provides data preprocessing and applies GLMMs for DEGs’ inference. The preprocessing allows efficient remotion of genes that could impact the inference. Also, the computational and biological validation of DEGRE has shown to be promising in identifying possible DEGs in experiments derived from complex experimental designs. This tool may help handle random effects on individuals in the inference of DEGs and presents a potential for discovering new interesting DEGs for further biological investigation.
Collapse
Affiliation(s)
- Douglas Terra Machado
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | - Yasmmin Côrtes Martins
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
28
|
Suarez LM, Diaz-Del Cerro E, Felix J, Gonzalez-Sanchez M, Ceprian N, Guerra-Perez N, G Novelle M, Martinez de Toda I, De la Fuente M. Sex differences in neuroimmunoendocrine communication. Involvement on longevity. Mech Ageing Dev 2023; 211:111798. [PMID: 36907251 DOI: 10.1016/j.mad.2023.111798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Endocrine, nervous, and immune systems work coordinately to maintain the global homeostasis of the organism. They show sex differences in their functions that, in turn, contribute to sex differences beyond reproductive function. Females display a better control of the energetic metabolism and improved neuroprotection and have more antioxidant defenses and a better inflammatory status than males, which is associated with a more robust immune response than that of males. These differences are present from the early stages of life, being more relevant in adulthood and influencing the aging trajectory in each sex and may contribute to the different life lifespan between sexes.
Collapse
Affiliation(s)
- Luz M Suarez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain.
| | - Estefania Diaz-Del Cerro
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Judith Felix
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica Gonzalez-Sanchez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Noemi Ceprian
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Natalia Guerra-Perez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Marta G Novelle
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain
| | - Irene Martinez de Toda
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica De la Fuente
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain.
| |
Collapse
|
29
|
Guma E, Beauchamp A, Liu S, Levitis E, Clasen LS, Torres E, Blumenthal J, Lalonde F, Qiu LR, Hrncir H, MacKenzie-Graham A, Yang X, Arnold AP, Lerch JP, Raznahan A. A Cross-Species Neuroimaging Study of Sex Chromosome Dosage Effects on Human and Mouse Brain Anatomy. J Neurosci 2023; 43:1321-1333. [PMID: 36631267 PMCID: PMC9987571 DOI: 10.1523/jneurosci.1761-22.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/12/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
All eutherian mammals show chromosomal sex determination with contrasting sex chromosome dosages (SCDs) between males (XY) and females (XX). Studies in transgenic mice and humans with sex chromosome trisomy (SCT) have revealed direct SCD effects on regional mammalian brain anatomy, but we lack a formal test for cross-species conservation of these effects. Here, we develop a harmonized framework for comparative structural neuroimaging and apply this to systematically profile SCD effects on regional brain anatomy in both humans and mice by contrasting groups with SCT (XXY and XYY) versus XY controls. Total brain size was substantially altered by SCT in humans (significantly decreased by XXY and increased by XYY), but not in mice. Robust and spatially convergent effects of XXY and XYY on regional brain volume were observed in humans, but not mice, when controlling for global volume differences. However, mice do show subtle effects of XXY and XYY on regional volume, although there is not a general spatial convergence in these effects within mice or between species. Notwithstanding this general lack of conservation in SCT effects, we detect several brain regions that show overlapping effects of XXY and XYY both within and between species (cerebellar, parietal, and orbitofrontal cortex), thereby nominating high priority targets for future translational dissection of SCD effects on the mammalian brain. Our study introduces a generalizable framework for comparative neuroimaging in humans and mice and applies this to achieve a cross-species comparison of SCD effects on the mammalian brain through the lens of SCT.SIGNIFICANCE STATEMENT Sex chromosome dosage (SCD) affects neuroanatomy and risk for psychopathology in humans. Performing mechanistic studies in the human brain is challenging but possible in mouse models. Here, we develop a framework for cross-species neuroimaging analysis and use this to show that an added X- or Y-chromosome significantly alters human brain anatomy but has muted effects in the mouse brain. However, we do find evidence for conserved cross-species impact of an added chromosome in the fronto-parietal cortices and cerebellum, which point to regions for future mechanistic dissection of sex chromosome dosage effects on brain development.
Collapse
Affiliation(s)
- Elisa Guma
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Antoine Beauchamp
- Mouse Imaging Centre, Toronto, Ontario M5T 3H7, Canada
- The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Siyuan Liu
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Elizabeth Levitis
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Liv S. Clasen
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Erin Torres
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Jonathan Blumenthal
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Francois Lalonde
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| | - Lily R. Qiu
- Mouse Imaging Centre, Toronto, Ontario M5T 3H7, Canada
- The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | - Haley Hrncir
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095
| | - Allan MacKenzie-Graham
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095
| | - Arthur P. Arnold
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095
| | - Jason P. Lerch
- Mouse Imaging Centre, Toronto, Ontario M5T 3H7, Canada
- The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Armin Raznahan
- Section on Developmental Neurogenomics, Human Genetics Branch, National Institute of Mental Health, Bethesda, 20892, Maryland
| |
Collapse
|
30
|
Brown RE. Sex Differences in Neurodevelopment and Its Disorders. NEURODEVELOPMENTAL PEDIATRICS 2023:179-212. [DOI: 10.1007/978-3-031-20792-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
31
|
Colby AE, DeCasien AR, Cooper EB, Higham JP. Greater variability in rhesus macaque ( Macaca mulatta) endocranial volume among males than females. Proc Biol Sci 2022; 289:20220728. [PMID: 36350207 PMCID: PMC9653222 DOI: 10.1098/rspb.2022.0728] [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: 04/14/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2023] Open
Abstract
The greater male variability (GMV) hypothesis proposes that traits are more variable among males than females, and is supported by numerous empirical studies. Interestingly, GMV is also observed for human brain size and internal brain structure, a pattern which may have implications for sex-biased neurological and psychiatric conditions. A better understanding of neuroanatomical variability in non-human primates may illuminate whether certain species are appropriate models for these conditions. Here, we tested for sex differences in the variability of endocranial volume (ECV, a proxy for brain size) in a sample of 542 rhesus macaques (Macaca mulatta) from a large pedigreed free-ranging population. We also examined the components of phenotypic variance (additive genetic and residual variance) to tease apart the potential drivers of sex differences in variability. Our results suggest that males exhibit more variable ECVs, and that this pattern reflects either balancing/disruptive selection on male behaviour (associated with alternative male mating strategies) or sex chromosome effects (associated with mosaic patterns of X chromosome gene expression in females), rather than extended neurodevelopment among males. This represents evidence of GMV for brain size in a non-human primate species and highlights the potential of rhesus macaques as a model for sex-biased brain-based disorders.
Collapse
Affiliation(s)
- Abigail E. Colby
- Department of Anthropology, New York University, New York, NY, USA
- Department of Anthropology and Archaeology, University of Calgary, Calgary, AB, Canada
| | - Alex R. DeCasien
- Department of Anthropology, New York University, New York, NY, USA
- New York Consortium in Evolutionary Primatology, New York, NY, USA
- Section on Developmental Neurogenomics, National Institutes of Health, Bethesda, MD, USA
| | - Eve B. Cooper
- Department of Anthropology, New York University, New York, NY, USA
- New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - James P. Higham
- Department of Anthropology, New York University, New York, NY, USA
- New York Consortium in Evolutionary Primatology, New York, NY, USA
| |
Collapse
|
32
|
She J, Du M, Xu Z, Jin Y, Li Y, Zhang D, Tao C, Chen J, Wang J, Yang E. The landscape of hervRNAs transcribed from human endogenous retroviruses across human body sites. Genome Biol 2022; 23:231. [PMID: 36329469 PMCID: PMC9632151 DOI: 10.1186/s13059-022-02804-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Human endogenous retroviruses (HERVs), the remnants of ancient retroviruses, account for 8% of the human genome, but most have lost their transcriptional abilities under physiological conditions. However, mounting evidence shows that several expressed HERVs do exert biological functions. Here, we systematically characterize physiologically expressed HERVs and examine whether they may give insight into the molecular fundamentals of human development and disease. RESULTS We systematically identify 13,889 expressed HERVs across normal body sites and demonstrate that they are expressed in body site-specific patterns and also by sex, ethnicity, and age. Analyzing cis-ERV-related quantitative trait loci, we find that 5435 hervRNAs are regulated by genetic variants. Combining this with a genome-wide association study, we elucidate that the dysregulation of expressed HERVs might be associated with various complex diseases, particularly neurodegenerative and psychiatric diseases. We further find that physiologically activated hervRNAs are associated with histone modifications rather than DNA demethylation. CONCLUSIONS Our results present a locus-specific landscape of physiologically expressed hervRNAs, which represent a hidden layer of genetic architecture in development and disease.
Collapse
Affiliation(s)
- Jianqi She
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, 100191, China
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Minghao Du
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, 100191, China
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Zhanzhan Xu
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yueqi Jin
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, 100191, China
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yu Li
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Daoning Zhang
- Peking University First Hospital, Beijing, 100034, China
| | - Changyu Tao
- Department of Human Anatomy, Histology & Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing, 102206, China
| | - Jiadong Wang
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ence Yang
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of China, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, 100191, China.
- Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Chinese Institute for Brain Research, Beijing, 102206, China.
- Taizhou Medical New & Hi-tech Industrial Development Zone, Jiangsu, 225326, China.
| |
Collapse
|
33
|
Cabrera Zapata LE, Garcia-Segura LM, Cambiasso MJ, Arevalo MA. Genetics and Epigenetics of the X and Y Chromosomes in the Sexual Differentiation of the Brain. Int J Mol Sci 2022; 23:ijms232012288. [PMID: 36293143 PMCID: PMC9603441 DOI: 10.3390/ijms232012288] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/27/2022] Open
Abstract
For many decades to date, neuroendocrinologists have delved into the key contribution of gonadal hormones to the generation of sex differences in the developing brain and the expression of sex-specific physiological and behavioral phenotypes in adulthood. However, it was not until recent years that the role of sex chromosomes in the matter started to be seriously explored and unveiled beyond gonadal determination. Now we know that the divergent evolutionary process suffered by X and Y chromosomes has determined that they now encode mostly dissimilar genetic information and are subject to different epigenetic regulations, characteristics that together contribute to generate sex differences between XX and XY cells/individuals from the zygote throughout life. Here we will review and discuss relevant data showing how particular X- and Y-linked genes and epigenetic mechanisms controlling their expression and inheritance are involved, along with or independently of gonadal hormones, in the generation of sex differences in the brain.
Collapse
Affiliation(s)
- Lucas E. Cabrera Zapata
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
| | | | - María Julia Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba 5016, Argentina
- Cátedra de Biología Celular, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba 5000, Argentina
- Correspondence: (M.J.C.); (M.A.A.)
| | - Maria Angeles Arevalo
- Instituto Cajal (IC), Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: (M.J.C.); (M.A.A.)
| |
Collapse
|
34
|
Pallier PN, Ferrara M, Romagnolo F, Ferretti MT, Soreq H, Cerase A. Chromosomal and environmental contributions to sex differences in the vulnerability to neurological and neuropsychiatric disorders: Implications for therapeutic interventions. Prog Neurobiol 2022; 219:102353. [PMID: 36100191 DOI: 10.1016/j.pneurobio.2022.102353] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/22/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
Neurological and neuropsychiatric disorders affect men and women differently. Multiple sclerosis, Alzheimer's disease, anxiety disorders, depression, meningiomas and late-onset schizophrenia affect women more frequently than men. By contrast, Parkinson's disease, autism spectrum condition, attention-deficit hyperactivity disorder, Tourette's syndrome, amyotrophic lateral sclerosis and early-onset schizophrenia are more prevalent in men. Women have been historically under-recruited or excluded from clinical trials, and most basic research uses male rodent cells or animals as disease models, rarely studying both sexes and factoring sex as a potential source of variation, resulting in a poor understanding of the underlying biological reasons for sex and gender differences in the development of such diseases. Putative pathophysiological contributors include hormones and epigenetics regulators but additional biological and non-biological influences may be at play. We review here the evidence for the underpinning role of the sex chromosome complement, X chromosome inactivation, and environmental and epigenetic regulators in sex differences in the vulnerability to brain disease. We conclude that there is a pressing need for a better understanding of the genetic, epigenetic and environmental mechanisms sustaining sex differences in such diseases, which is critical for developing a precision medicine approach based on sex-tailored prevention and treatment.
Collapse
Affiliation(s)
- Patrick N Pallier
- Blizard Institute, Centre for Neuroscience, Surgery and Trauma, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| | - Maria Ferrara
- Institute of Psychiatry, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy; Department of Psychiatry, Yale University, School of Medicine, New Haven, CT, United States; Women's Brain Project (WBP), Switzerland
| | - Francesca Romagnolo
- Institute of Psychiatry, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | | | - Hermona Soreq
- The Edmond and Lily Safra Center of Brain Science, The Hebrew University of Jerusalem, 9190401, Israel
| | - Andrea Cerase
- EMBL-Rome, Via Ramarini 32, 00015 Monterotondo, RM, Italy; Blizard Institute, Centre for Genomics and Child Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK; Department of Biology, University of Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy.
| |
Collapse
|
35
|
Munds RA, Cooper EB, Janiak MC, Lam LG, DeCasien AR, Bauman Surratt S, Montague MJ, Martinez MI, Research Unit CB, Kawamura S, Higham JP, Melin AD. Variation and heritability of retinal cone ratios in a free-ranging population of rhesus macaques. Evolution 2022; 76:1776-1789. [PMID: 35790204 PMCID: PMC9544366 DOI: 10.1111/evo.14552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/03/2022] [Accepted: 05/12/2022] [Indexed: 01/22/2023]
Abstract
A defining feature of catarrhine primates is uniform trichromacy-the ability to distinguish red (long; L), green (medium; M), and blue (short; S) wavelengths of light. Although the tuning of photoreceptors is conserved, the ratio of L:M cones in the retina is variable within and between species, with human cone ratios differing from other catarrhines. Yet, the sources and structure of variation in cone ratios are poorly understood, precluding a broader understanding of color vision variability. Here, we report a large-scale study of a pedigreed population of rhesus macaques (Macaca mulatta). We collected foveal RNA and analyzed opsin gene expression using cDNA and estimated additive genetic variance of cone ratios. The average L:M ratio and standard error was 1.03:1 ± 0.02. There was no age effect, and genetic contribution to variation was negligible. We found marginal sex effects with females having larger ratios than males. S cone ratios (0.143:1 ± 0.002) had significant genetic variance with a heritability estimate of 43% but did not differ between sexes or age groups. Our results contextualize the derived human condition of L-cone dominance and provide new information about the heritability of cone ratios and variation in primate color vision.
Collapse
Affiliation(s)
- Rachel A. Munds
- Department of Anthropology and ArchaeologyUniversity of CalgaryCalgaryABT2N 1N4Canada
| | - Eve B. Cooper
- Department of AnthropologyNew York UniversityNew YorkNew York10003,New York Consortium in Evolutionary PrimatologyNew YorkNew York10460
| | - Mareike C. Janiak
- Department of Anthropology and ArchaeologyUniversity of CalgaryCalgaryABT2N 1N4Canada,Department of AnthropologyNew York UniversityNew YorkNew York10003,School of Science, Engineering and EnvironmentUniversity of SalfordSalfordM5 4NTUnited Kingdom
| | - Linh Gia Lam
- Department of Anthropology and ArchaeologyUniversity of CalgaryCalgaryABT2N 1N4Canada
| | - Alex R. DeCasien
- Department of AnthropologyNew York UniversityNew YorkNew York10003,New York Consortium in Evolutionary PrimatologyNew YorkNew York10460,Section on Developmental NeurogenomicsNational Institute of Mental HealthBethesdaMaryland20892
| | | | - Michael J. Montague
- Department of NeuroscienceUniversity of PennsylvaniaPhiladelphiaPennsylvania19104
| | - Melween I. Martinez
- Caribbean Primate Research CenterUniversity of Puerto RicoSan JuanPuerto Rico00936
| | | | - Shoji Kawamura
- Department of Integrated BiosciencesUniversity of TokyoKashiwa277‐8562Japan
| | - James P. Higham
- Department of AnthropologyNew York UniversityNew YorkNew York10003,New York Consortium in Evolutionary PrimatologyNew YorkNew York10460
| | - Amanda D. Melin
- Department of Anthropology and ArchaeologyUniversity of CalgaryCalgaryABT2N 1N4Canada,Department of Medical GeneticsUniversity of CalgaryCalgaryABT2N 1N4Canada,Alberta Children's Hospital Research InstituteUniversity of CalgaryCalgaryABT2N 1N4Canada
| |
Collapse
|
36
|
DeCasien AR, Guma E, Liu S, Raznahan A. Sex differences in the human brain: a roadmap for more careful analysis and interpretation of a biological reality. Biol Sex Differ 2022; 13:43. [PMID: 35883159 PMCID: PMC9327177 DOI: 10.1186/s13293-022-00448-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
The presence, magnitude, and significance of sex differences in the human brain are hotly debated topics in the scientific community and popular media. This debate is largely fueled by studies containing strong, opposing conclusions: either little to no evidence exists for sex differences in human neuroanatomy, or there are small-to-moderate differences in the size of certain brain regions that are highly reproducible across cohorts (even after controlling for sex differences in average brain size). Our Commentary uses the specific comparison between two recent large-scale studies that adopt these opposing views-namely the review by Eliot and colleagues (2021) and the direct analysis of ~ 40k brains by Williams and colleagues (2021)-in an effort to clarify this controversy and provide a framework for conducting this research. First, we review observations that motivate research on sex differences in human neuroanatomy, including potential causes (evolutionary, genetic, and environmental) and effects (epidemiological and clinical evidence for sex-biased brain disorders). We also summarize methodological and empirical support for using structural MRI to investigate such patterns. Next, we outline how researchers focused on sex differences can better specify their study design (e.g., how sex was defined, if and how brain size was adjusted for) and results (by e.g., distinguishing sexual dimorphisms from sex differences). We then compare the different approaches available for studying sex differences across a large number of individuals: direct analysis, meta-analysis, and review. We stress that reviews do not account for methodological differences across studies, and that this variation explains many of the apparent inconsistencies reported throughout recent reviews (including the work by Eliot and colleagues). For instance, we show that amygdala volume is consistently reported as male-biased in studies with sufficient sample sizes and appropriate methods for brain size correction. In fact, comparing the results from multiple large direct analyses highlights small, highly reproducible sex differences in the volume of many brain regions (controlling for brain size). Finally, we describe best practices for the presentation and interpretation of these findings. Care in interpretation is important for all domains of science, but especially so for research on sex differences in the human brain, given the existence of broad societal gender-biases and a history of biological data being used justify sexist ideas. As such, we urge researchers to discuss their results from simultaneously scientific and anti-sexist viewpoints.
Collapse
Affiliation(s)
- Alex R DeCasien
- Section On Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA.
| | - Elisa Guma
- Section On Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Siyuan Liu
- Section On Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| | - Armin Raznahan
- Section On Developmental Neurogenomics, National Institute of Mental Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Bakker J. The role of steroid hormones in the sexual differentiation of the human brain. J Neuroendocrinol 2022; 34:e13050. [PMID: 34708466 DOI: 10.1111/jne.13050] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/03/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022]
Abstract
Widespread sex differences in human brain structure and function have been reported. Research on animal models has demonstrated that sex differences in brain and behavior are induced by steroid hormones during specific, hormone sensitive, developmental periods. It was shown that typical male neural and behavioral characteristics develop under the influence of testosterone, mostly acting during perinatal development. By contrast, typical female neural and behavioral characteristics may actually develop under the influence of estradiol during a specific prepubertal period. This review provides an overview of our current knowledge on the role of steroid hormones in the sexual differentiation of the human brain. Both clinical and neuroimaging data obtained in patients with altered androgen levels/actions (i.e., congenital adrenal hyperplasia or complete androgen insensitivity syndrome [CAIS]), point to an important role of (prenatal) androgens in inducing typical male neural and psychosexual characteristics in humans. In contrast to rodents, there appears to be no obvious role for estrogens in masculinizing the human brain. Furthermore, data from CAIS also suggest a contribution of sex chromosome genes to the development of the human brain. The final part of this review is dedicated to a brief discussion of gender incongruence, also known as gender dysphoria, which has been associated with an altered or less pronounced sexual differentiation of the brain.
Collapse
Affiliation(s)
- Julie Bakker
- Neuroendocrinology, GIGA Neurosciences, Liège University, Liege, Belgium
| |
Collapse
|
38
|
Tronson NC. Puberty reverses sex differences in learning. Nat Neurosci 2022; 25:134-135. [PMID: 35087247 DOI: 10.1038/s41593-021-00986-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Parra-Montes de Oca MA, Sotelo-Rivera I, Gutiérrez-Mata A, Charli JL, Joseph-Bravo P. Sex Dimorphic Responses of the Hypothalamus-Pituitary-Thyroid Axis to Energy Demands and Stress. Front Endocrinol (Lausanne) 2021; 12:746924. [PMID: 34745011 PMCID: PMC8565401 DOI: 10.3389/fendo.2021.746924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/15/2021] [Indexed: 12/16/2022] Open
Abstract
The hypothalamus-pituitary-thyroid-axis (HPT) is one of the main neuroendocrine axes that control energy expenditure. The activity of hypophysiotropic thyrotropin releasing hormone (TRH) neurons is modulated by nutritional status, energy demands and stress, all of which are sex dependent. Sex dimorphism has been associated with sex steroids whose concentration vary along the life-span, but also to sex chromosomes that define not only sexual characteristics but the expression of relevant genes. In this review we describe sex differences in basal HPT axis activity and in its response to stress and to metabolic challenges in experimental animals at different stages of development, as well as some of the limited information available on humans. Literature review was accomplished by searching in Pubmed under the following words: "sex dimorphic" or "sex differences" or "female" or "women" and "thyrotropin" or "thyroid hormones" or "deiodinases" and "energy homeostasis" or "stress". The most representative articles were discussed, and to reduce the number of references, selected reviews were cited.
Collapse
Affiliation(s)
| | | | | | | | - Patricia Joseph-Bravo
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| |
Collapse
|
40
|
Fang H, Deng X, Disteche CM. X-factors in human disease: Impact of gene content and dosage regulation. Hum Mol Genet 2021; 30:R285-R295. [PMID: 34387327 DOI: 10.1093/hmg/ddab221] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/13/2022] Open
Abstract
The gene content of the X and Y chromosomes has dramatically diverged during evolution. The ensuing dosage imbalance within the genome of males and females has led to unique chromosome-wide regulatory mechanisms with significant and sex-specific impacts on X-linked gene expression. X inactivation or silencing of most genes on one X chromosome chosen at random in females profoundly affects the manifestation of X-linked diseases, as males inherit a single maternal allele, while females express maternal and paternal alleles in a mosaic manner. An additional complication is the existence of genes that escape X inactivation and thus are ubiquitously expressed from both alleles in females. The mosaic nature of X-linked gene expression and the potential for escape can vary between individuals, tissues, cell types, and stages of life. Our understanding of the specialized nature of X-linked genes and of the multilayer epigenetic regulation that influence their expression throughout the organism has been helped by molecular studies conducted by tissue-specific and single-cell-specific approaches. In turn, the definition of molecular events that control X silencing has helped develop new approaches for the treatment of some X-linked disorders. This review focuses on the peculiarities of the X chromosome genetic content and epigenetic regulation in shaping the manifestation of congenital and acquired X-linked disorders in a sex-specific manner.
Collapse
Affiliation(s)
- He Fang
- Department of Laboratory Medicine and Pathology
| | | | - Christine M Disteche
- Department of Laboratory Medicine and Pathology.,Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
41
|
Affiliation(s)
- Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,UNC Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
42
|
Chowen JA, Garcia-Segura LM. Role of glial cells in the generation of sex differences in neurodegenerative diseases and brain aging. Mech Ageing Dev 2021; 196:111473. [PMID: 33766745 DOI: 10.1016/j.mad.2021.111473] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, and IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain.
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
43
|
Moisan MP. Sexual Dimorphism in Glucocorticoid Stress Response. Int J Mol Sci 2021; 22:ijms22063139. [PMID: 33808655 PMCID: PMC8003420 DOI: 10.3390/ijms22063139] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Chronic stress is encountered in our everyday life and is thought to contribute to a number of diseases. Many of these stress-related disorders display a sex bias. Because glucocorticoid hormones are the main biological mediator of chronic stress, researchers have been interested in understanding the sexual dimorphism in glucocorticoid stress response to better explain the sex bias in stress-related diseases. Although not yet demonstrated for glucocorticoid regulation, sex chromosomes do influence sex-specific biology as soon as conception. Then a transient rise in testosterone start to shape the male brain during the prenatal period differently to the female brain. These organizational effects are completed just before puberty. The cerebral regions implicated in glucocorticoid regulation at rest and after stress are thereby impacted in a sex-specific manner. After puberty, the high levels of all gonadal hormones will interact with glucocorticoid hormones in specific crosstalk through their respective nuclear receptors. In addition, stress occurring early in life, in particular during the prenatal period and in adolescence will prime in the long-term glucocorticoid stress response through epigenetic mechanisms, again in a sex-specific manner. Altogether, various molecular mechanisms explain sex-specific glucocorticoid stress responses that do not exclude important gender effects in humans.
Collapse
|