1
|
Ågmo A. Androgen receptors and sociosexual behaviors in mammals: The limits of generalization. Neurosci Biobehav Rev 2024; 157:105530. [PMID: 38176634 DOI: 10.1016/j.neubiorev.2023.105530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Circulating testosterone is easily aromatized to estradiol and reduced to dihydrotestosterone in target tissues and elsewhere in the body. Thus, the actions of testosterone can be mediated either by the estrogen receptors, the androgen receptor or by simultaneous action at both receptors. To determine the role of androgens acting at the androgen receptor, we need to eliminate actions at the estrogen receptors. Alternatively, actions at the androgen receptor itself can be eliminated. In the present review, I will analyze the specific role of androgen receptors in male and female sexual behavior as well as in aggression. Some comments about androgen receptors and social recognition are also made. It will be shown that there are important differences between species, even between strains within a species, concerning the actions of the androgen receptor on the behaviors mentioned. This fact makes generalizations from one species to another or from one strain to another very risky. The existence of important species differences is often ignored, leading to many misunderstandings and much confusion.
Collapse
Affiliation(s)
- Anders Ågmo
- Department of Psychology, University of Tromsø, Norway.
| |
Collapse
|
2
|
Craft RM, Sewell CM, Taylor TM, Vo MS, Delevich K, Morgan MM. Impact of continuous testosterone exposure on reproductive physiology, activity, and pain-related behavior in young adult female rats. Horm Behav 2024; 158:105469. [PMID: 38091929 DOI: 10.1016/j.yhbeh.2023.105469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/02/2023] [Accepted: 12/04/2023] [Indexed: 02/05/2024]
Abstract
Testosterone may reduce pain in cisgender women and transgender men. Rodents can provide a useful model for investigating physiological effects of hormone therapy. To this end, continuous-release testosterone or blank (placebo) capsules were implanted s.c. into young adult female rats, and three weeks later rats were either ovariectomized or sham-ovariectomized. Testosterone treatment that mimicked previously reported endogenous levels in males eliminated estrous cycling and decreased uterine weight. Testosterone also significantly increased body weight and suppressed the increases in daily wheel running observed in placebo controls over time. Subsequent ovariectomy or sham-ovariectomy decreased wheel running in all groups, but testosterone-treated rats recovered significantly more quickly than did placebo-treated rats. Neither testosterone nor ovariectomy significantly altered hindpaw mechanical threshold. Two weeks after sham/ovariectomy surgery, injection of Complete Freund Adjuvant (CFA) into one hindpaw reduced wheel running and mechanical threshold in all groups; running significantly decreased from the first to second day after CFA in testosterone- but not in placebo-treated rats. Morphine 1.0 but not 3.2 mg/kg increased CFA-suppressed wheel running similarly in all groups, whereas both doses of morphine increased CFA-suppressed mechanical threshold. These data suggest that weeks-long testosterone treatment with or without ovariectomy may provide a useful physiological model of testosterone therapy as used in human gender transition. Although testosterone administered at levels similar to those in gonadally intact males tended to hasten female rats' recovery from surgery, it did not decrease maximal pain-related behaviors after surgery or hindpaw inflammatory insult, nor did it alter opioid antinociception.
Collapse
Affiliation(s)
- Rebecca M Craft
- Department of Psychology, Washington State University, Pullman and Vancouver, WA, United States of America.
| | - Christyne M Sewell
- Department of Psychology, Washington State University, Pullman and Vancouver, WA, United States of America
| | - Tessa M Taylor
- Department of Psychology, Washington State University, Pullman and Vancouver, WA, United States of America
| | - Mai Suong Vo
- Department of Psychology, Washington State University, Pullman and Vancouver, WA, United States of America
| | - Kristen Delevich
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, United States of America
| | - Michael M Morgan
- Department of Psychology, Washington State University, Pullman and Vancouver, WA, United States of America
| |
Collapse
|
3
|
Williams JS, Fattori MR, Honeyborne IR, Ritz SA. Considering hormones as sex- and gender-related factors in biomedical research: Challenging false dichotomies and embracing complexity. Horm Behav 2023; 156:105442. [PMID: 37913648 DOI: 10.1016/j.yhbeh.2023.105442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 11/03/2023]
Abstract
The inclusion of sex and gender considerations in biomedicine has been increasing in light of calls from research and funding agencies, governmental bodies, and advocacy groups to direct research attention to these issues. Although the inclusion of both female and male participants is often an important element, overreliance on a female-male binary tends to oversimplify the interactions between sex- and gender-related factors and health, and runs a risk of being influenced by cultural stereotypes about sex and gender. When biomedical researchers are examining how hormones associated with gender and sex may influence pathways of interest, it is of crucial importance to approach this work with a critical lens on the rhetoric used, and in ways that acknowledge the complexity of hormone physiology. Here, we document the ways in which discourses around sex, gender and hormones shape our scientific thinking and practice in biomedical research, and review how the existing scientific knowledge about hormones reflects a complex and dynamic reality that is often not reflected outside of specialist niches of hormone biology. Where biomedical scientists take up sex- and gender-associated hormones as a way of addressing sex and gender considerations, it is valuable for us to bring a critical lens to the rhetoric and discourses used, to employ a sex contextualist approach in designing experimentation, and be rigorous and reflexive about the approaches used in analysis and interpretation of data. These strategies will allow us to design experimentation that goes beyond binaries, and grapples more directly with the material intricacies of sex, gender, and hormones.
Collapse
Affiliation(s)
| | - Michelle R Fattori
- Health Sciences Education Program, McMaster University, Hamilton, Ontario, Canada
| | - Isabella R Honeyborne
- Bachelor of Health Sciences (Honours) Program, McMaster University, Hamilton, Ontario, Canada
| | - Stacey A Ritz
- Department of Pathology & Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Lahimer M, Abou Diwan M, Montjean D, Cabry R, Bach V, Ajina M, Ben Ali H, Benkhalifa M, Khorsi-Cauet H. Endocrine disrupting chemicals and male fertility: from physiological to molecular effects. Front Public Health 2023; 11:1232646. [PMID: 37886048 PMCID: PMC10598475 DOI: 10.3389/fpubh.2023.1232646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
The deleterious effects of chemical or non-chemical endocrine disruptors (EDs) on male fertility potential is well documented but still not fully elucidated. For example, the detection of industrial chemicals' metabolites in seminal plasma and follicular fluid can affect efficiency of the gametogenesis, the maturation and competency of gametes and has guided scientists to hypothesize that endocrine disrupting chemicals (EDCs) may disrupt hormonal homoeostasis by leading to a wide range of hormonal control impairments. The effects of EDCs exposure on reproductive health are highly dependent on factors including the type of EDCs, the duration of exposure, individual susceptibility, and the presence of other co-factors. Research and scientists continue to study these complex interactions. The aim of this review is to summarize the literature to better understand the potential reproductive health risks of EDCs in France.
Collapse
Affiliation(s)
- Marwa Lahimer
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, CHU Sud, Amiens, France
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
- Exercise Physiology and Physiopathology: from Integrated to Molecular “Biology, Medicine and Health” (Code: LR19ES09), Sousse, Tunisia
| | - Maria Abou Diwan
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
| | - Debbie Montjean
- Fertilys, Centres de Fertilité, Laval and Brossard, QC, Canada
| | - Rosalie Cabry
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, CHU Sud, Amiens, France
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
| | - Véronique Bach
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
| | - Mounir Ajina
- Service of Reproductive Medicine, University Hospital Farhat Hached, Sousse, Tunisia
| | - Habib Ben Ali
- Laboratory Histology Embryology, Faculty of Medicine Sousse, University of Sousse, Sousse, Tunisia
| | - Moncef Benkhalifa
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, CHU Sud, Amiens, France
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
| | - Hafida Khorsi-Cauet
- ART and Reproductive Biology Laboratory, University Hospital and School of Medicine, CHU Sud, Amiens, France
- PERITOX-(UMR-I 01), UPJV/INERIS, UPJV, CURS, Chemin du Thil, Amiens, France
| |
Collapse
|
5
|
Olasore HSA, Oyedeji TA, Olawale MO, Ogundele OI, Faleti JOO. Relationship between testosterone-estradiol ratio and some anthropometric and metabolic parameters among Nigerian men. Metabol Open 2023; 18:100249. [PMID: 37396673 PMCID: PMC10313505 DOI: 10.1016/j.metop.2023.100249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 07/04/2023] Open
Abstract
Background Alterations in sex hormone levels are implicated in the regulation of metabolic processes in men. In recent years, the prevalence of metabolic disorders, such as obesity, insulin resistance, and type 2 diabetes, has risen in Nigeria. In men, these disorders may be associated with the ratio of serum testosterone to estradiol levels. Therefore, we investigated the relationship between the testosterone-estradiol (T/E2) ratio, anthropometry, and metabolic parameters in Nigerian men. Method Eighty-five adult men were recruited for this study. Participants' data such as age, weight, height, BMI, and waist circumference were collected. Plasma total testosterone and estradiol levels, as well as metabolic parameters such as fasting blood sugar, creatinine, urea, HDL cholesterol, total cholesterol, and triglycerides levels, were determined. The data were analyzed using SPSS version 25 software. Results Anthropometric parameters such as weight, height, BMI, and waist circumference showed a negative correlation with plasma T/E2 (r = -0.265, -0.288, -0.106, -0.204; p = 0.007, 0.004, 0.167, 0.061 respectively). However, the T/E2 ratio showed a positive correlation with the metabolic parameters such as fasting blood sugar, HDL cholesterol levels, plasma creatinine, and urea (r = 0.219, 0.096, 0.992, 0.152; p = 0.022, 0.192, <0.001, 0.082 respectively), while there were negative correlations with total cholesterol and triglycerides levels (r = -0.200, -0.083; p = 0.034, 0.226 respectively). Conclusion These findings show that there are significant correlations between the T/E2 ratio and weight, height, fasting blood sugar, creatinine, and urea, while there are no significant correlations between T/E2 ratio and BMI, waist circumference, HDL-cholesterol, and triglycerides.
Collapse
Affiliation(s)
- Holiness Stephen Adedeji Olasore
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Tolulope Adejoke Oyedeji
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | - Matthew Olamide Olawale
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| | | | - Joseph Ogo-Oluwa Faleti
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of the University of Lagos, Idi Araba, Lagos, Nigeria
| |
Collapse
|
6
|
Lu Y, Qin L, Wei Y, Mo X, Tang X, Liu Q, Liu S, Zhang J, Xu M, Wei C, Huang S, Lin Y, Luo T, Mai T, Gou R, Zhang Z, Cai J, Qin J. Association between barium exposed, CYP19A1 and central obesity: A cross-sectional study in rural China. J Trace Elem Med Biol 2023; 78:127170. [PMID: 37075568 DOI: 10.1016/j.jtemb.2023.127170] [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: 11/11/2022] [Revised: 03/04/2023] [Accepted: 04/03/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND obesity is a major risk factor for many metabolic diseases such as diabetes and cardiometabolic diseases. This study aimed to evaluate the association of plasma and urinary barium concentrations, CYP19A1 gene polymorphisms, and their interaction with central obesity in a rural Chinese population. METHODS restricted cubic spline model was used to explore the dose-response relationship between barium and the risk of developing central obesity and waist circumference; logistic regression model was used to assess the association between barium, CYP19A1 gene polymorphisms and their interaction with central obesity. RESULTS the results of the restricted cubic spline model showed that plasma barium concentration was linearly associated with the risk of developing central obesity and non-linearly associated with waist circumference. Logistic regression analysis showed that participants with Q4 plasma barium concentration exhibited a higher risk of central obesity compared to participants with Q1 barium concentration; participants carrying the rs10046-AA gene exhibited a lower risk of central obesity than those carrying the rs10046-G(GG+GA) gene; participants carrying the rs10046-GA genotype showed 1.754 times higher risk of central obesity than those carrying rs10046-GG+AA genotype. There was a significant interaction between plasma barium and CYP19A1 gene polymorphism on central obesity. CONCLUSION the development of central obesity was associated with plasma barium and CYP19A1.
Collapse
Affiliation(s)
- Yufu Lu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Lidong Qin
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Yanfei Wei
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Xiaoting Mo
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Xu Tang
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Qiumei Liu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Shuzhen Liu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Junling Zhang
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Min Xu
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Chunmei Wei
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Shenxiang Huang
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Yinxia Lin
- School of Public Health, Guangxi Medical University, Shuangyong Road No.22, Nanning 530021, Guangxi, China
| | - Tingyu Luo
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Tingyu Mai
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Ruoyu Gou
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China
| | - Zhiyong Zhang
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China; Guangxi key laboratory of Environmental Exposomics and Entire Lifecycle Health, China
| | - Jiansheng Cai
- School of Public Health, Guilin Medical University, 20 Lequn Road, Guilin, Guangxi, China; Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, Guangxi, China.
| | - Jian Qin
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education, Guangxi Medical University, Nanning 530021, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
7
|
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]
|
8
|
Moruno-Manchon J, Noh B, McCullough L. Sex-biased autophagy as a potential mechanism mediating sex differences in ischemic stroke outcome. Neural Regen Res 2023; 18:31-37. [PMID: 35799505 PMCID: PMC9241419 DOI: 10.4103/1673-5374.340406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Stroke is the second leading cause of death and a major cause of disability worldwide, and biological sex is an important determining factor in stroke incidence and pathology. From childhood through adulthood, men have a higher incidence of stroke compared with women. Abundant research has confirmed the beneficial effects of estrogen in experimental ischemic stroke but genetic factors such as the X-chromosome complement can also play an important role in determining sex differences in stroke. Autophagy is a self-degrading cellular process orchestrated by multiple core proteins, which leads to the engulfment of cytoplasmic material and degradation of cargo after autophagy vesicles fuse with lysosomes or endosomes. The levels and the activity of components of these signaling pathways and of autophagy-related proteins can be altered during ischemic insults. Ischemic stroke activates autophagy, however, whether inhibiting autophagy after stroke is beneficial in the brain is still under a debate. Autophagy is a potential mechanism that may contribute to differences in stroke progression between the sexes. Furthermore, the effects of manipulating autophagy may also differ between the sexes. Mechanisms that regulate autophagy in a sex-dependent manner in ischemic stroke remain unexplored. In this review, we summarize clinical and pre-clinical evidence for sex differences in stroke. We briefly introduce the autophagy process and summarize the effects of gonadal hormones in autophagy in the brain and discuss X-linked genes that could potentially regulate brain autophagy. Finally, we review pre-clinical studies that address the mechanisms that could mediate sex differences in brain autophagy after stroke.
Collapse
|
9
|
Peng Q, Yan Y, Yan H, Xie G, Shi L, Wen Y, Chang Q. Association between CYP19A1 rs6493497 and rs936306 polymorphisms and depression susceptibility in the Chinese population. Biomark Med 2022; 16:1171-1179. [PMID: 36628958 DOI: 10.2217/bmm-2022-0370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aim: The CYP19A1 gene encodes the key aromatase for estrogen biosynthesis, and this study aimed to explore the relationship between CYP19A1 rs6493497 and rs936306 polymorphisms and depression risk. Methods: CYP19A1 rs6493497 and rs936306 genotyping was performed on 502 depression patients and 504 healthy controls. Results: In the general population, no significant association was observed between the CYP19A1 rs6493497 variant and depression, whereas that CYP19A1 rs936306 variant significantly reduced depression risk in the recessive model. In subgroup analysis, a significant association of the CYP19A1 rs6493497 variant with reduced depression risk was found in males aged 46-65 in the genotype, dominant and additive models. Conclusion: The CYP19A1 rs936306 variant may reduce depression risk, and the rs6493497 variant is associated with decreased depression risk in males aged 46-65.
Collapse
Affiliation(s)
- Qiuju Peng
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Yan
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huacheng Yan
- Department of Infectious Disease Prevention & Control, Center for Disease Control & Prevention of Southern Theatre Command, Guangzhou, 510507, China
| | - Guibo Xie
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Shi
- Department of Pharmacy, General Hospital of Southern Theatre Command, Guangzhou, 510010, China
| | - Yuguan Wen
- Department of Pharmacy, Guangzhou Brain Hospital, Guangzhou, 510370, China
| | - Qingxian Chang
- Department of Gynecology & Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
10
|
Yan Y, Wang X, Chaput D, Shin MK, Koh Y, Gan L, Pieper AA, Woo JAA, Kang DE. X-linked ubiquitin-specific peptidase 11 increases tauopathy vulnerability in women. Cell 2022; 185:3913-3930.e19. [PMID: 36198316 PMCID: PMC9588697 DOI: 10.1016/j.cell.2022.09.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/31/2022] [Accepted: 08/31/2022] [Indexed: 01/26/2023]
Abstract
Although women experience significantly higher tau burden and increased risk for Alzheimer's disease (AD) than men, the underlying mechanism for this vulnerability has not been explained. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that X-linked ubiquitin specific peptidase 11 (USP11) augments pathological tau aggregation via tau deubiquitination initiated at lysine-281. Removal of ubiquitin provides access for enzymatic tau acetylation at lysines 281 and 274. USP11 escapes complete X-inactivation, and female mice and people both exhibit higher USP11 levels than males. Genetic elimination of usp11 in a tauopathy mouse model preferentially protects females from acetylated tau accumulation, tau pathology, and cognitive impairment. USP11 levels also strongly associate positively with tau pathology in females but not males. Thus, inhibiting USP11-mediated tau deubiquitination may provide an effective therapeutic opportunity to protect women from increased vulnerability to AD and other tauopathies.
Collapse
Affiliation(s)
- Yan Yan
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Dale Chaput
- Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Min-Kyoo Shin
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Yeojung Koh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Cleveland, Louis Stokes Cleveland VA Medical Center, OH, USA; Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jung-A A Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA.
| | - David E Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Louis Strokes Cleveland VA Medical Center, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA.
| |
Collapse
|
11
|
Chinn GA, Duong K, Horovitz TR, Russell JMS, Sall JW. Testosterone is Sufficient to Impart Susceptibility to Isoflurane Neurotoxicity in Female Neonatal Rats. J Neurosurg Anesthesiol 2022; 34:429-436. [PMID: 34127616 PMCID: PMC8671561 DOI: 10.1097/ana.0000000000000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Volatile anesthetic exposure during development leads to long-term cognitive deficits in rats which are dependent on age and sex. Female rats are protected relative to male rats for the same exposure on postnatal day 7. Here we test our hypothesis that androgens can modulate chloride cotransporter expression to alter the susceptibility to neurotoxicity from GABAergic drugs using female rats with exogenous testosterone exposure. METHODS Female rats were injected with testosterone (100 μg/animal) or vehicle on postnatal days 1 to 6. On postnatal day 7, the animals were randomized to either isoflurane exposure or sham. Spatial memory was assessed with the Barnes maze starting on postnatal day 41. Western blots were run from testosterone treated postnatal day 7 animals to measure levels of chloride cotransporters sodium-potassium-chloride symporter (NKCC1) and chloride-potassium symporter 5 (KCC2). RESULTS Exogenous testosterone modulated isoflurane anesthetic neurotoxicity in female rats based on poor performance in the probe trial of the Barnes Maze. By contrast, females with vehicle and isoflurane exposure were able to differentiate the goal position. These behavioral differences corresponded to differences in the protein levels of NKCC1 and KCC2 after exogenous testosterone exposure, with NKCC1 increasing ( P <0.001) and KCC2 decreasing ( P =0.003) relative to female controls. CONCLUSIONS The expression of chloride cotransporters, NKCC1 and KCC2, is altered by testosterone in female rats and corresponds to a cognitive deficit after isoflurane exposure. This confirms the role of androgens in perinatal anesthetic neurotoxicity and supports our hypothesis that the developing GABAergic system plays a critical role in the underlying mechanism.
Collapse
Affiliation(s)
- Gregory A Chinn
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Katrina Duong
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Tal R Horovitz
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Jennifer M Sasaki Russell
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Jeffrey W Sall
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| |
Collapse
|
12
|
Orsini CA, Truckenbrod LM, Wheeler AR. Regulation of sex differences in risk-based decision making by gonadal hormones: Insights from rodent models. Behav Processes 2022; 200:104663. [PMID: 35661794 PMCID: PMC9893517 DOI: 10.1016/j.beproc.2022.104663] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 04/22/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023]
Abstract
Men and women differ in their ability to evaluate options that vary in their rewards and the risks that are associated with these outcomes. Most studies have shown that women are more risk averse than men and that gonadal hormones significantly contribute to this sex difference. Gonadal hormones can influence risk-based decision making (i.e., risk taking) by modulating the neurobiological substrates underlying this cognitive process. Indeed, estradiol, progesterone and testosterone modulate activity in the prefrontal cortex, amygdala and nucleus accumbens associated with reward and risk-related information. The use of animal models of decision making has advanced our understanding of the intersection between the behavioral, neural and hormonal mechanisms underlying sex differences in risk taking. This review will outline the current state of this literature, identify the current gaps in knowledge and suggest the neurobiological mechanisms by which hormones regulate risky decision making. Collectively, this knowledge can be used to understand the potential consequences of significant hormonal changes, whether endogenously or exogenously induced, on risk-based decision making as well as the neuroendocrinological basis of neuropsychiatric diseases that are characterized by impaired risk taking, such as substance use disorder and schizophrenia.
Collapse
Affiliation(s)
- Caitlin A. Orsini
- Department of Psychology, University of Texas at Austin, Austin, TX, USA,Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA,Correspondence to: Department of Psychology & Neurology, Waggoner Center for Alcohol and Addiction Research, 108 E. Dean Keaton St., Stop A8000, Austin, TX 78712, USA. (C.A. Orsini)
| | - Leah M. Truckenbrod
- Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| | - Alexa-Rae Wheeler
- Department of Neurology, University of Texas at Austin, Austin, TX, USA,Waggoner Center for Alcohol and Addiction Research, University of Texas at Austin, Austin, TX, USA,Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
13
|
Lazar V, Holban AM, Curutiu C, Ditu LM. Modulation of Gut Microbiota by Essential Oils and Inorganic Nanoparticles: Impact in Nutrition and Health. Front Nutr 2022; 9:920413. [PMID: 35873448 PMCID: PMC9305160 DOI: 10.3389/fnut.2022.920413] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Microbiota plays a crucial role in human health and disease; therefore, the modulation of this complex and yet widely unexplored ecosystem is a biomedical priority. Numerous antibacterial alternatives have been developed in recent years, imposed by the huge problem of antibioresistance, but also by the people demand for natural therapeutical products without side effects, as dysbiosis, cyto/hepatotoxicity. Current studies are focusing mainly in the development of nanoparticles (NPs) functionalized with herbal and fruit essential oils (EOs) to fight resistant pathogens. This is due to their increased efficiency against susceptible, multidrug resistant and biofilm embedded microorganisms. They are also studied because of their versatile properties, size and possibility to ensure a targeted administration and a controlled release of bioactive substances. Accordingly, an increasing number of studies addressing the effects of functional nanoparticles and plant products on microbial pathogens has been observed. Regardless the beneficial role of EOs and NPs in the treatment of infectious diseases, concerns regarding their potential activity against human microbiota raised constantly in recent years. The main focus of current research is on gut microbiota (GM) due to well documented metabolic and immunological functions of gut microbes. Moreover, GM is constantly exposed to micro- and nano-particles, but also plant products (including EOs). Because of the great diversity of both microbiota and chemical antimicrobial alternatives (i.e., nanomaterials and EOs), here we limit our discussion on the interactions of gut microbiota, inorganic NPs and EOs. Impact of accidental exposure caused by ingestion of day care products, foods, atmospheric particles and drugs containing nanoparticles and/or fruit EOs on gut dysbiosis and associated diseases is also dissected in this paper. Current models developed to investigate mechanisms of dysbiosis after exposure to NPs/EOs and perspectives for identifying factors driving EOs functionalized NPs dysbiosis are reviewed.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Alina-Maria Holban
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Laboratory of Microbiology, Research Institute of the University of Bucharest, Bucharest, Romania
- *Correspondence: Alina-Maria Holban
| | - Carmen Curutiu
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Lia Mara Ditu
- Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Laboratory of Microbiology, Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
14
|
Spool JA, Bergan JF, Remage-Healey L. A neural circuit perspective on brain aromatase. Front Neuroendocrinol 2022; 65:100973. [PMID: 34942232 PMCID: PMC9667830 DOI: 10.1016/j.yfrne.2021.100973] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/23/2022]
Abstract
This review explores the role of aromatase in the brain as illuminated by a set of conserved network-level connections identified in several vertebrate taxa. Aromatase-expressing neurons are neurochemically heterogeneous but the brain regions in which they are found are highly-conserved across the vertebrate lineage. During development, aromatase neurons have a prominent role in sexual differentiation of the brain and resultant sex differences in behavior and human brain diseases. Drawing on literature primarily from birds and rodents, we delineate brain regions that express aromatase and that are strongly interconnected, and suggest that, in many species, aromatase expression essentially defines the Social Behavior Network. Moreover, in several cases the inputs to and outputs from this core Social Behavior Network also express aromatase. Recent advances in molecular and genetic tools for neuroscience now enable in-depth and taxonomically diverse studies of the function of aromatase at the neural circuit level.
Collapse
Affiliation(s)
- Jeremy A Spool
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Joseph F Bergan
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Luke Remage-Healey
- Center for Neuroendocrine Studies, Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, United States.
| |
Collapse
|
15
|
Royal W, Bryant J, Davis H, Guo M. Cigarette smoke and nicotine effects on behavior in HIV transgenic rats. Behav Brain Res 2022; 417:113591. [PMID: 34551347 PMCID: PMC9107950 DOI: 10.1016/j.bbr.2021.113591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 01/26/2023]
Abstract
HIV-related neurocognitive impairment can be worsened by cigarette smoking and be more severe in women. Therefore, we analyzed the effects of sex on behavioral function in HIV transgenic (Tg) rats that were exposed to either nicotine alone, to smoke from either nicotine-containing or nicotine-free cigarettes, or non-exposed. The animals were then assessed on the open field test for the total distance traveled and for the fraction of the total distance traveled and the total time spent in the center of the field, and the results then compared to WT rats subjected to the same exposures and testing. Higher total distances indicate greater locomotor activity and a higher center field measures imply a lower anxiety state. Total distances were overall higher for female and for Tg rats exposed to nicotine-free CS. Also, the total distance and both center field measures were overall higher for female rats in the control and nicotine-free CS-exposed groups. This was observed specifically for WT females as compared to WT males and, for the center field measures, for WT females as compared to Tg males. No genotype or sex-related differences were found for rats in the nicotine-free cigarette smoke (CS) and nicotine-containing CS exposed groups. Therefore, nicotine exposure did not impact genotype- and sex-related differences in motor responses and anxiety levels that were found in the control state. However, exposure to the non-nicotine components of CS resulted in locomotor activation in the presence of the HIV genes and was anxiogenic in WT and Tg male animals.
Collapse
Affiliation(s)
- Walter Royal
- Morehouse School of Medicine Department of Neurobiology and Neuroscience Institute, Georgia; Atlanta VA Medical Center, USA; University of Maryland School of Medicine, USA.
| | - Joseph Bryant
- University of Maryland School Institute of Human Virology, USA; University of Maryland School of Medicine, USA
| | - Harry Davis
- University of Maryland School Institute of Human Virology, USA; University of Maryland School of Medicine, USA
| | - Ming Guo
- University of Maryland School of Medicine, USA
| |
Collapse
|
16
|
Yu H, Niu Y, Jia G, Liang Y, Chen B, Sun R, Wang M, Huang S, Zeng J, Lu J, Li L, Guo X, Yao P. Maternal diabetes-mediated RORA suppression in mice contributes to autism-like offspring through inhibition of aromatase. Commun Biol 2022; 5:51. [PMID: 35027651 PMCID: PMC8758718 DOI: 10.1038/s42003-022-03005-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/23/2021] [Indexed: 01/31/2023] Open
Abstract
Retinoic acid-related orphan receptor alpha (RORA) suppression is associated with autism spectrum disorder (ASD) development, although the mechanism remains unclear. In this study, we aim to investigate the potential effect and mechanisms of RORA suppression on autism-like behavior (ALB) through maternal diabetes-mediated mouse model. Our in vitro study in human neural progenitor cells shows that transient hyperglycemia induces persistent RORA suppression through oxidative stress-mediated epigenetic modifications and subsequent dissociation of octamer-binding transcription factor 3/4 from the RORA promoter, subsequently suppressing the expression of aromatase and superoxide dismutase 2. The in vivo mouse study shows that prenatal RORA deficiency in neuron-specific RORA null mice mimics maternal diabetes-mediated ALB; postnatal RORA expression in the amygdala ameliorates, while postnatal RORA knockdown mimics, maternal diabetes-mediated ALB in offspring. In addition, RORA mRNA levels in peripheral blood mononuclear cells decrease to 34.2% in ASD patients (n = 121) compared to the typically developing group (n = 118), and the related Receiver Operating Characteristic curve shows good sensitivity and specificity with a calculated 84.1% of Area Under the Curve for ASD diagnosis. We conclude that maternal diabetes contributes to ALB in offspring through suppression of RORA and aromatase, RORA expression in PBMC could be a potential marker for ASD screening. Hong Yu, Yanbin Niu, Guohua Jia et al. integrate in vitro, in vivo, and human experiments to examine a link between RORA expression on autism-like behavior. Their results suggest that maternal diabetes may contribute to autism-like behavior via RORA suppression.
Collapse
Affiliation(s)
- Hong Yu
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China
| | - Yanbin Niu
- Teachers College, Columbia University, New York, NY, 10027, USA
| | - Guohua Jia
- Hainan Women and Children's Medical Center, Haikou, 570206, P. R. China
| | - Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen Mental Health Center, Shenzhen, 518020, P. R. China
| | - Baolin Chen
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China
| | - Ruoyu Sun
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China
| | - Min Wang
- Hainan Women and Children's Medical Center, Haikou, 570206, P. R. China
| | - Saijun Huang
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China
| | - Jiaying Zeng
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China
| | - Jianpin Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen Mental Health Center, Shenzhen, 518020, P. R. China
| | - Ling Li
- Hainan Women and Children's Medical Center, Haikou, 570206, P. R. China.
| | - Xiaoling Guo
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China.
| | - Paul Yao
- Department of Pediatrics, Foshan Maternity and Child Health Care Hospital, Foshan, 528041, P. R. China. .,Hainan Women and Children's Medical Center, Haikou, 570206, P. R. China.
| |
Collapse
|
17
|
Ho TC, Gifuni AJ, Gotlib IH. Psychobiological risk factors for suicidal thoughts and behaviors in adolescence: a consideration of the role of puberty. Mol Psychiatry 2022; 27:606-623. [PMID: 34117365 PMCID: PMC8960417 DOI: 10.1038/s41380-021-01171-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/03/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023]
Abstract
Suicide is the second leading cause of death among adolescents. While clinicians and researchers have begun to recognize the importance of considering multidimensional factors in understanding risk for suicidal thoughts and behaviors (STBs) during this developmental period, the role of puberty has been largely ignored. In this review, we contend that the hormonal events that occur during puberty have significant effects on the organization and development of brain systems implicated in the regulation of social stressors, including amygdala, hippocampus, striatum, medial prefrontal cortex, orbitofrontal cortex, and anterior cingulate cortex. Guided by previous experimental work in adults, we also propose that the influence of pubertal hormones and social stressors on neural systems related to risk for STBs is especially critical to consider in adolescents with a neurobiological sensitivity to hormonal changes. Furthermore, facets of the pubertal transition, such as pubertal timing, warrant deeper investigation and may help us gain a more comprehensive understanding of sex differences in the neurobiological and psychosocial mechanisms underlying adolescent STBs. Ultimately, advancing our understanding of the pubertal processes that contribute to suicide risk will improve early detection and facilitate the development of more effective, sex-specific, psychiatric interventions for adolescents.
Collapse
Affiliation(s)
- Tiffany C. Ho
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA ,grid.266102.10000 0001 2297 6811Department of Psychiatry and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA USA
| | - Anthony J. Gifuni
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA ,grid.14709.3b0000 0004 1936 8649Psychiatry Department and Douglas Mental Health University Institute, McGill University, Montréal, QC Canada
| | - Ian H. Gotlib
- grid.168010.e0000000419368956Department of Psychology, Stanford University, Stanford, CA USA
| |
Collapse
|
18
|
PET imaging of brain aromatase in humans and rhesus monkeys by 11C-labeled cetrozole analogs. Sci Rep 2021; 11:23623. [PMID: 34880350 PMCID: PMC8654920 DOI: 10.1038/s41598-021-03063-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/22/2021] [Indexed: 11/08/2022] Open
Abstract
Aromatase is an estrogen synthetic enzyme that plays important roles in brain functions. To quantify aromatase expression in the brain by positron emission tomography (PET), we had previously developed [11C]cetrozole, which showed high specificity and affinity. To develop more efficient PET tracer(s) for aromatase imaging, we synthesized three analogs of cetrozole. We synthesized meta-cetrozole, nitro-cetrozole, and iso-cetrozole, and prepared the corresponding 11C-labeled tracers. The inhibitory activities of these three analogs toward aromatase were evaluated using marmoset placenta, and PET imaging of brain aromatase was performed using the 11C-labeled tracers in monkeys. The most promising analog in the monkey study, iso-cetrozole, was evaluated in the human PET study. The highest to lowest inhibitory activity of the analogs toward aromatase in the microsomal fraction from marmoset placenta was in the following order: iso-cetrozole, nitro-cetrozole, cetrozole, and meta-cetrozole. This order showed good agreement with the order of the binding potential (BP) of each 11C-labeled analog to aromatase in the rhesus monkey brain. A human PET study using [11C]iso-analog showed a similar distribution pattern of binding as that of [11C]cetrozole. The time-activity curves showed that elimination of [11C]iso-cetrozole from brain tissue was faster than that of 11C-cetrozole, indicating more rapid metabolism of [11C]iso-cetrozole. [11C]Cetrozole has preferable metabolic stability for brain aromatase imaging in humans, although [11C]iso-cetrozole might also be useful to measure aromatase level in living human brain because of its high binding potential.
Collapse
|
19
|
Arnold ML, Saijo K. Estrogen Receptor β as a Candidate Regulator of Sex Differences in the Maternal Immune Activation Model of ASD. Front Mol Neurosci 2021; 14:717411. [PMID: 34531723 PMCID: PMC8438209 DOI: 10.3389/fnmol.2021.717411] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
Interestingly, more males are diagnosed with autism spectrum disorder (ASD) than females, yet the mechanism behind this difference is unclear. Genes on the sex chromosomes and differential regulation by sex steroid hormones and their receptors are both candidate mechanisms to explain this sex-dependent phenotype. Nuclear receptors (NRs) are a large family of transcription factors, including sex hormone receptors, that mediate ligand-dependent transcription and may play key roles in sex-specific regulation of immunity and brain development. Infection during pregnancy is known to increase the probability of developing ASD in humans, and a mouse model of maternal immune activation (MIA), which is induced by injecting innate immune stimulants into pregnant wild-type mice, is commonly used to study ASD. Since this model successfully recaptures the behavioral phenotypes and male bias observed in ASD, we will discuss the potential role of sex steroid hormones and their receptors, especially focusing on estrogen receptor (ER)β, in MIA and how this signaling may modulate transcription and subsequent inflammation in myeloid-lineage cells to contribute to the etiology of this neurodevelopmental disorder.
Collapse
Affiliation(s)
- Madeline L Arnold
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States.,Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
20
|
Brandt N, Vierk R, Fester L, Anstötz M, Zhou L, Heilmann LF, Kind S, Steffen P, Rune GM. Sex-specific Difference of Hippocampal Synaptic Plasticity in Response to Sex Neurosteroids. Cereb Cortex 2021; 30:2627-2641. [PMID: 31800024 DOI: 10.1093/cercor/bhz265] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/23/2019] [Accepted: 09/17/2019] [Indexed: 12/14/2022] Open
Abstract
Numerous studies provide increasing evidence, which supports the ideas that every cell in the brain of males may differ from those in females due to differences in sex chromosome complement as well as in response to hormonal effects. In this study, we address the question as to whether actions of neurosteroids, thus steroids, which are synthesized and function within the brain, contribute to sex-specific hippocampal synaptic plasticity. We have previously shown that predominantly in the female hippocampus, does inhibition of the conversion of testosterone to estradiol affect synaptic transmission. In this study, we show that testosterone and its metabolite dihydrotestosterone are essential for hippocampal synaptic transmission specifically in males. This also holds true for the density of mushroom spines and of spine synapses. We obtained similar sex-dependent results using primary hippocampal cultures of male and female animals. Since these cultures originated from perinatal animals, our findings argue for sex-dependent differentiation of hippocampal neurons regarding their responsiveness to sex neurosteroids up to birth, which persist during adulthood. Hence, our in vitro findings may point to a developmental effect either directly induced by sex chromosomes or indirectly by fetal testosterone secretion during the perinatal critical period, when developmental sexual priming takes place.
Collapse
Affiliation(s)
- Nicola Brandt
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Ricardo Vierk
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lars Fester
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Max Anstötz
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lepu Zhou
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lukas F Heilmann
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Simon Kind
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Paul Steffen
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
21
|
Abaffy T, Matsunami H. 19-hydroxy Steroids in the Aromatase Reaction: Review on Expression and Potential Functions. J Endocr Soc 2021; 5:bvab050. [PMID: 34095690 PMCID: PMC8169043 DOI: 10.1210/jendso/bvab050] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Indexed: 12/05/2022] Open
Abstract
Scientific evidence related to the aromatase reaction in various biological processes spanning from mid-1960 to today is abundant; however, as our analytical sensitivity increases, a new look at the old chemical reaction is necessary. Here, we review an irreversible aromatase reaction from the substrate androstenedione. It proceeds in 3 consecutive steps. In the first 2 steps, 19-hydroxy steroids are produced. In the third step, estrone is produced. They can dissociate from the enzyme complex and either accumulate in tissues or enter the blood. In this review, we want to highlight the potential importance of these 19-hydroxy steroids in various physiological and pathological conditions. We focus primarily on 19-hydroxy steroids, and in particular on the 19-hydroxyandrostenedione produced by the incomplete aromatase reaction. Using a PubMed database and the search term “aromatase reaction,” 19-hydroxylation of androgens and steroid measurements, we detail the chemistry of the aromatase reaction and list previous and current methods used to measure 19-hydroxy steroids. We present evidence of the existence of 19-hydroxy steroids in brain tissue, ovaries, testes, adrenal glands, prostate cancer, as well as during pregnancy and parturition and in Cushing’s disease. Based on the available literature, a potential involvement of 19-hydroxy steroids in the brain differentiation process, sperm motility, ovarian function, and hypertension is suggested and warrants future research. We hope that with the advancement of highly specific and sensitive analytical methods, future research into 19-hydroxy steroids will be encouraged, as much remains to be learned and discovered.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| |
Collapse
|
22
|
Lyu Z, Ghoshdastidar S, Rekha KR, Suresh D, Mao J, Bivens N, Kannan R, Joshi T, Rosenfeld CS, Upendran A. Developmental exposure to silver nanoparticles leads to long term gut dysbiosis and neurobehavioral alterations. Sci Rep 2021; 11:6558. [PMID: 33753813 PMCID: PMC7985313 DOI: 10.1038/s41598-021-85919-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Due to their antimicrobial properties, silver nanoparticles (AgNPs) are used in a wide range of consumer products that includes topical wound dressings, coatings for biomedical devices, and food-packaging to extend the shelf-life. Despite their beneficial antimicrobial effects, developmental exposure to such AgNPs may lead to gut dysbiosis and long-term health consequences in exposed offspring. AgNPs can cross the placenta and blood–brain-barrier to translocate in the brain of offspring. The underlying hypothesis tested in the current study was that developmental exposure of male and female mice to AgNPs disrupts the microbiome–gut–brain axis. To examine for such effects, C57BL6 female mice were exposed orally to AgNPs at a dose of 3 mg/kg BW or vehicle control 2 weeks prior to breeding and throughout gestation. Male and female offspring were tested in various mazes that measure different behavioral domains, and the gut microbial profiles were surveyed from 30 through 120 days of age. Our study results suggest that developmental exposure results in increased likelihood of engaging in repetitive behaviors and reductions in resident microglial cells. Echo-MRI results indicate increased body fat in offspring exposed to AgNPs exhibit. Coprobacillus spp., Mucispirillum spp., and Bifidobacterium spp. were reduced, while Prevotella spp., Bacillus spp., Planococcaceae, Staphylococcus spp., Enterococcus spp., and Ruminococcus spp. were increased in those developmentally exposed to NPs. These bacterial changes were linked to behavioral and metabolic alterations. In conclusion, developmental exposure of AgNPs results in long term gut dysbiosis, body fat increase and neurobehavioral alterations in offspring.
Collapse
Affiliation(s)
- Zhen Lyu
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65212, USA
| | - Shreya Ghoshdastidar
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, 65211, USA
| | - Karamkolly R Rekha
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
| | - Dhananjay Suresh
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, 65211, USA
| | - Jiude Mao
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65212, USA.,MU Institute of Data Science and Informatics, University of Missouri, Columbia, MO, 65212, USA
| | - Nathan Bivens
- DNA Core Facility, University of Missouri, Columbia, MO, 65212, USA
| | - Raghuraman Kannan
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, 65211, USA.,Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
| | - Trupti Joshi
- Department of Electrical Engineering and Computer Science, University of Missouri, Columbia, MO, 65212, USA. .,Department of Health Management and Informatics, University of Missouri, Columbia, MO, 65212, USA. .,MU Institute of Data Science and Informatics, University of Missouri, Columbia, MO, 65212, USA. .,Bond Life Sciences Center, University of Missouri, Columbia, MO, 65212, USA.
| | - Cheryl S Rosenfeld
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, 65212, USA. .,MU Institute of Data Science and Informatics, University of Missouri, Columbia, MO, 65212, USA. .,Bond Life Sciences Center, University of Missouri, Columbia, MO, 65212, USA. .,Genetics Area Program, University of Missouri, Columbia, MO, 65212, USA. .,Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, 65212, USA. .,Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO, 65212, USA.
| | - Anandhi Upendran
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO, 65212, USA. .,MU-Institute of Clinical and Translational Science, University of Missouri, Columbia, MO, 65212, USA.
| |
Collapse
|
23
|
Addo KA, Palakodety N, Fry RC. Acetaminophen Modulates the Expression of Steroidogenesis-Associated Genes and Estradiol Levels in Human Placental JEG-3 Cells. Toxicol Sci 2021; 179:44-52. [PMID: 33098425 PMCID: PMC8599781 DOI: 10.1093/toxsci/kfaa160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Acetaminophen is the only medication recommended for pain and fever management during pregnancy. However, studies have reported an association between in utero acetaminophen and neurocognitive disorders later in life. Additionally, acetaminophen has been shown to have endocrine disrupting properties altering hormones critical for normal fetal development. As the placenta is an endocrine organ that produces hormones for fetal development, any attempts to elucidate the mechanism underlying in utero acetaminophen and birth outcomes must also focus on the placenta. The present study set out to examine the effect of acetaminophen on mRNA expression, protein expression, and hormone synthesis in placental JEG-3 cells. The analysis focused on genes involved in steroidogenesis and acetaminophen metabolism as well those with known roles as nuclear receptors and transporters. The results highlight that at high concentrations, acetaminophen reduced the gene expression of aromatase (CYP19A1) and type 1 3β-hydroxysteroid dehydrogenase (HSD3B1), and increased the expression of 17β-hydroxysteroid dehydrogenase (HSD17B1). Additionally, acetaminophen at high concentrations also reduced the protein expression of aromatase (CYP19A1). These effects were accompanied by a significant dose-dependent decrease in estradiol secretion. Estradiol plays an important role in the development of reproductive organs and the brain of the developing fetus. This study highlights the potential for acetaminophen to interfere with hormone regulation during pregnancy and underscores the need for additional studies aimed at understanding the endocrine disruption activity of acetaminophen during fetal development.
Collapse
Affiliation(s)
- Kezia A Addo
- Curriculum in Toxicology and Environmental Medicine, School of Medicine
- Department of Environmental Sciences and Engineering
- ICF International, Durham, North Carolina
| | | | - Rebecca C Fry
- Curriculum in Toxicology and Environmental Medicine, School of Medicine
- Department of Environmental Sciences and Engineering
- Institute for Environmental Health Solutions, Gilling School of Global Public Health, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
24
|
Varshney MK, Yu NYL, Katayama S, Li X, Liu T, Wu WF, Töhönen V, Krjutškov K, Kere J, Fan X, Inzunza J, Gustafsson JÅ, Nalvarte I. Motor Function Deficits in the Estrogen Receptor Beta Knockout Mouse: Role on Excitatory Neurotransmission and Myelination in the Motor Cortex. Neuroendocrinology 2021; 111:27-44. [PMID: 31991411 DOI: 10.1159/000506162] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/25/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Male estrogen receptor beta (ERβ) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERβ loss have not been investigated. OBJECTIVE The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERβ loss on OL differentiation were confirmed in vitro. CONCLUSION ERβ is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.
Collapse
Affiliation(s)
| | - Nancy Yiu-Lin Yu
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Shintaro Katayama
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Xin Li
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Wan-Fu Wu
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Virpi Töhönen
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Kaarel Krjutškov
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Competence Center on Health Technologies, Tartu, Estonia
- Folkhälsan Research Institute, Helsinki, and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Juha Kere
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Folkhälsan Research Institute, Helsinki, and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - José Inzunza
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ivan Nalvarte
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden,
| |
Collapse
|
25
|
Qian L, Qi S, Zhang J, Duan M, Schlenk D, Jiang J, Wang C. Exposure to Boscalid Induces Reproductive Toxicity of Zebrafish by Gender-Specific Alterations in Steroidogenesis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:14275-14287. [PMID: 33138376 DOI: 10.1021/acs.est.0c02871] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Boscalid is a succinate dehydrogenase inhibitor fungicide and is frequently detected in surface water. Due to the frequent detection of boscalid, we evaluated its impact on the reproduction of adult zebrafish following a 21 d exposure to 0, 0.01, 0.1, and 1.0 mg/L. Following exposure to boscalid, the fertility of female zebrafish and fertilization rate of spawning eggs were reduced in a concentration-dependent manner up to a respective 87% and 20% in the highest concentration. A significant 16% reduction in the percentage of late vitellogenic oocytes was noted in ovaries, and a significant 74% reduction in the percentage of spermatids in testis was also observed after treatment with 1.0 mg/L. 17β-Estradiol (E2) concentrations decreased significantly in females (34% decrease) but significantly increased in males (15% increase) following 1.0 mg/L boscalid treatment. The expression of genes (such as era, er2b, cyp19a, and cyp19b) related to the hypothalamus-pituitary-gonad-liver (HPGL) axis was significantly altered and positively correlated with E2 concentrations in female and male zebrafish (p < 0.05). Molecular docking results revealed that the binding modes between boscalid and target proteins (ER and CYP19) of zebrafish were similar to that of the reference compounds and the target proteins. The binding energies indicate that boscalid may have a weak estrogen-like binding effect or CYP19 inhibition, potentially altering the HPGL axis, thereby reducing E2 concentrations and fecundity in females. In contrast, boscalid caused significant induction of E2 steroidogenesis and subsequent feminization of gonads in males, indicating gender-specific adverse outcome pathways.
Collapse
Affiliation(s)
- Le Qian
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Suzhen Qi
- Risk Assessment Laboratory for Bee Product Quality and Safety of Ministry of Agriculture, Institute of Agricultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, People's Republic of China
| | - Jie Zhang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Manman Duan
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, Riverside, California 92521, United States
| | - Jiazhen Jiang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Chengju Wang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| |
Collapse
|
26
|
Alia-Klein N, Preston-Campbell RN, Kim SW, Pareto D, Logan J, Wang GJ, Moeller SJ, Fowler JS, Biegon A. Human Cognitive Ability Is Modulated by Aromatase Availability in the Brain in a Sex-Specific Manner. Front Neurosci 2020; 14:565668. [PMID: 33192252 PMCID: PMC7604391 DOI: 10.3389/fnins.2020.565668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/14/2020] [Indexed: 11/18/2022] Open
Abstract
The enzyme aromatase catalyzes the final step in estrogen biosynthesis, converting testosterone to estradiol, and is expressed in the brain of all mammals. Estrogens are thought to be important for maintenance of cognitive function in women, whereas testosterone is thought to modulate cognitive abilities in men. Here, we compare differences in cognitive performance in relation to brain aromatase availability in healthy men and women. Twenty-seven healthy participants were administered tests of verbal learning and memory and perceptual/abstract reasoning. In vivo images of brain aromatase availability were acquired in this sample using positron emission tomography (PET) with the validated aromatase radiotracer [11C]vorozole. Regions of interest were placed bilaterally on the amygdala and thalamus where aromatase availability is highest in the human brain. Though cognitive performance and aromatase availability did not differ as a function of sex, higher availability of aromatase in the amygdala was associated with lower cognitive performance in men. No such relationship was found in women; and the corresponding regression slopes were significantly different between the sexes. Thalamic aromatase availability was not significantly correlated with cognitive performance in either sex. These findings suggest that the effects of brain aromatase on cognitive performance are both region- and sex-specific and may explain some of the normal variance seen in verbal and nonverbal cognitive abilities in men and women as well as sex differences in the trajectory of cognitive decline associated with Alzheimer’s disease.
Collapse
Affiliation(s)
- Nelly Alia-Klein
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Sung Won Kim
- National Institute on Alcohol and Alcohol Abuse, Bethesda, MD, United States
| | - Deborah Pareto
- Neuroradiology Unit, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Jean Logan
- New York University, Langone Medical Center, New York, NY, United States
| | - Gene-Jack Wang
- National Institute on Alcohol and Alcohol Abuse, Bethesda, MD, United States
| | - Scott J. Moeller
- Stony Brook University School of Medicine, Stony Brook, NY, United States
| | | | - Anat Biegon
- Stony Brook University School of Medicine, Stony Brook, NY, United States
- *Correspondence: Anat Biegon,
| |
Collapse
|
27
|
Shay DA, Welly RJ, Givan SA, Bivens N, Kanaley J, Marshall BL, Lubahn DB, Rosenfeld CS, Vieira-Potter VJ. Changes in nucleus accumbens gene expression accompany sex-specific suppression of spontaneous physical activity in aromatase knockout mice. Horm Behav 2020; 121:104719. [PMID: 32081742 PMCID: PMC7387966 DOI: 10.1016/j.yhbeh.2020.104719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 12/29/2022]
Abstract
Aromatase catalyzes conversion of testosterone to estradiol and is expressed in a variety of tissues, including the brain. Suppression of aromatase adversely affects metabolism and physical activity behavior, but mechanisms remain uncertain. The hypothesis tested herein was that whole body aromatase deletion would cause gene expression changes in the nucleus accumbens (NAc), a brain regulating motivated behaviors such as physical activity, which is suppressed with loss of estradiol. Metabolic and behavioral assessments were performed in male and female wild-type (WT) and aromatase knockout (ArKO) mice. NAc-specific differentially expressed genes (DEGs) were identified with RNAseq, and associations between the measured phenotypic traits were determined. Female ArKO mice had greater percent body fat, reduced spontaneous physical activity (SPA), consumed less energy, and had lower relative resting energy expenditure (REE) than WT females. Such differences were not observed in ArKO males. However, in both sexes, a top DEG was Pts, a gene encoding an enzyme necessary for catecholamine (e.g., dopamine) biosynthesis. In comparing male and female WT mice, top DEGs were related to sexual development/fertility, immune regulation, obesity, dopamine signaling, and circadian regulation. SPA correlated strongly with Per3, a gene regulating circadian function, thermoregulation, and metabolism (r = -0.64, P = .002), which also correlated with adiposity (r = 0.54, P = .01). In conclusion, aromatase ablation leads to gene expression changes in NAc, which may in turn result in reduced SPA and related metabolic abnormalities. These findings may have significance to post-menopausal women and those treated with an aromatase inhibitor.
Collapse
Affiliation(s)
- Dusti A Shay
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia 65211, MO, USA
| | - Rebecca J Welly
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia 65211, MO, USA
| | - Scott A Givan
- Informatics Research Core Facility, University of Missouri, Columbia 65211, MO, USA
| | - Nathan Bivens
- DNA Core Facility, University of Missouri, Columbia 65211, MO, USA
| | - Jill Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia 65211, MO, USA
| | - Brittney L Marshall
- Bond Life Sciences Center, University of Missouri, Columbia 65211, MO, USA; Biomedical Sciences, University of Missouri, Columbia 65211, MO, USA
| | - Dennis B Lubahn
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA; Department of Child Health, University of Missouri, Columbia, MO 65211, USA
| | - Cheryl S Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia 65211, MO, USA; Biomedical Sciences, University of Missouri, Columbia 65211, MO, USA; Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia 65211, MO, USA; MU Informatics Institute, University of Missouri, Columbia 65211, MO, USA
| | - Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia 65211, MO, USA.
| |
Collapse
|
28
|
Xiang D, Lu J, Wei C, Cai X, Wang Y, Liang Y, Xu M, Wang Z, Liu M, Wang M, Liang X, Li L, Yao P. Berberine Ameliorates Prenatal Dihydrotestosterone Exposure-Induced Autism-Like Behavior by Suppression of Androgen Receptor. Front Cell Neurosci 2020; 14:87. [PMID: 32327976 PMCID: PMC7161090 DOI: 10.3389/fncel.2020.00087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/23/2020] [Indexed: 01/03/2023] Open
Abstract
Many epidemiology studies have shown that maternal polycystic ovary syndrome (PCOS) results in a greater risk of autism spectrum disorders (ASD) development, although the detailed mechanism remains unclear. In this study, we aimed to investigate the potential mechanism and provide a possible treatment for PCOS-mediated ASD through three experiments: Experiment 1: real-time PCR and western blots were employed to measure gene expression in human neurons, and the luciferase reporter assay and chromatin immunoprecipitation (ChIP) was used to map the responsive elements on related gene promoters. Experiment 2: pregnant dams were prenatally exposed to dihydrotestosterone (DHT), androgen receptor (AR) knockdown (shAR) in the amygdala, or berberine (BBR), and the subsequent male offspring were used for autism-like behavior (ALB) assay followed by biomedical analysis, including gene expression, oxidative stress, and mitochondrial function. Experiment 3: the male offspring from prenatal DHT exposed dams were postnatally treated by either shAR or BBR, and the offspring were used for ALB assay followed by biomedical analysis. Our findings showed that DHT treatment suppresses the expression of estrogen receptor β (ERβ) and superoxide dismutase 2 (SOD2) through AR-mediated hypermethylation on the ERβ promoter, and BBR treatment suppresses AR expression through hypermethylation on the AR promoter. Prenatal DHT treatment induces ERβ suppression, oxidative stress and mitochondria dysfunction in the amygdala with subsequent ALB behavior in male offspring, and AR knockdown partly diminishes this effect. Furthermore, both prenatal and postnatal treatment of BBR partly restores prenatal DHT exposure-mediated ALB. In conclusion, DHT suppresses ERβ expression through the AR signaling pathway by hypermethylation on the ERβ promoter, and BBR restores this effect through AR suppression. Prenatal DHT exposure induces ALB in offspring through AR-mediated ERβ suppression, and both prenatal and postnatal treatment of BBR ameliorates this effect. We conclude that BBR ameliorates prenatal DHT exposure-induced ALB through AR suppression, this study may help elucidate the potential mechanism and identify a potential treatment through using BBR for PCOS-mediated ASD.
Collapse
Affiliation(s)
- Dongfang Xiang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianping Lu
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Chongxia Wei
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Xiaofan Cai
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Yongxia Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Liang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Mingtao Xu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zichen Wang
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China
| | - Min Liu
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Wang
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Xuefang Liang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Li
- Hainan Maternal and Child Health Hospital, Haikou, China
| | - Paul Yao
- Department of Child Psychiatry, Kangning Hospital of Shenzhen, Shenzhen, China.,Hainan Maternal and Child Health Hospital, Haikou, China
| |
Collapse
|
29
|
Delevich K, Okada NJ, Rahane A, Zhang Z, Hall CD, Wilbrecht L. Sex and Pubertal Status Influence Dendritic Spine Density on Frontal Corticostriatal Projection Neurons in Mice. Cereb Cortex 2020; 30:3543-3557. [PMID: 32037445 DOI: 10.1093/cercor/bhz325] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In humans, nonhuman primates, and rodents, the frontal cortices exhibit grey matter thinning and dendritic spine pruning that extends into adolescence. This maturation is believed to support higher cognition but may also confer psychiatric vulnerability during adolescence. Currently, little is known about how specific cell types in the frontal cortex mature or whether puberty plays a role in the maturation of some cell types but not others. Here, we used mice to characterize the spatial topography and adolescent development of cross-corticostriatal (cSTR) neurons that project through the corpus collosum to the dorsomedial striatum. We found that apical spine density on cSTR neurons in the medial prefrontal cortex decreased significantly between late juvenile (P29) and young adult time points (P60), with females exhibiting higher spine density than males at both ages. Adult males castrated prior to puberty onset had higher spine density compared to sham controls. Adult females ovariectomized before puberty onset showed greater variance in spine density measures on cSTR cells compared to controls, but their mean spine density did not significantly differ from sham controls. Our findings reveal that these cSTR neurons, a subtype of the broader class of intratelencephalic-type neurons, exhibit significant sex differences and suggest that spine pruning on cSTR neurons is regulated by puberty in male mice.
Collapse
Affiliation(s)
- Kristen Delevich
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Nana J Okada
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Ameet Rahane
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Zicheng Zhang
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Christopher D Hall
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| | - Linda Wilbrecht
- Department of Psychology, University of California, Berkeley, CA 94720, USA and.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA
| |
Collapse
|
30
|
Grigsby KB, Kovarik CM, Mao X, Booth FW. Medial preoptic estrogen receptor-beta blunts the estrogen receptor-alpha mediated increases in wheel-running behavior of female rats. Behav Brain Res 2020; 379:112341. [PMID: 31711895 DOI: 10.1016/j.bbr.2019.112341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/22/2019] [Accepted: 10/31/2019] [Indexed: 11/17/2022]
Abstract
Estrogens are believed to enhance rodent voluntary wheel-running through medial preoptic (mPOA) estrogen receptor α (ERα) signaling, with little role attributed to estrogen receptor β (ERβ). Systemic ERβ activation has been shown to mitigate ERα driven increases in wheel-running. Therefore, the present goal was to determine whether ERβ signaling in the mPOA plays a similar modulatory role over ERα. We utilized outbred wild-type (WT) and rats selectively bred for low voluntary running (LVR) behavior to address whether mPOA ERβ signaling blunts ERα driven wheel-running behavior and immediate-early gene (Fos, Zif268, and Homer1) mRNA induction. Further, we addressed baseline mPOA mRNA expressions and circulating 17β-estradiol levels between female WT and LVR rats. Following ovariectomy, WT rats reduced running behavior ∼40 %, with no effect in LVR rats. Intra-medial preoptic injection of the ERα-agonist propylpyrazoletriol (PPT) increased wheel-running ∼3.5-fold in WT rats, while injections of the ERβ-agonist diarylpropionitrile (DPN) or a combination of the two agonists had no effect. Similarly, ERα-agonism (PPT) increased Fos and Homer1 induction ∼3-fold in WT and LVR isolated mPOA neurons, with no effect of the ERβ-agonist DPN alone or in combination with PPT, suggesting medial-preoptic ERβ activity may blunt ERα signaling. LVR rats exhibited higher mPOA mRNA expressions of Esr1, Esr2 and Cyp19a1, lower normalized uterine wet weights and lower 17β-estradiol plasma levels compared to WT, suggesting their low running may be due to low circulating estrogen levels. Collectively, these findings highlight mPOA ERβ as a potential neuro-molecular modulator of the estrogenic control of wheel-running behavior.
Collapse
Affiliation(s)
- Kolter B Grigsby
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States.
| | - Cathleen M Kovarik
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Xuansong Mao
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, United States; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States; Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
31
|
Ancelin ML, Norton J, Canonico M, Scarabin PY, Ritchie K, Ryan J. Aromatase (CYP19A1) gene variants, sex steroid levels, and late-life depression. Depress Anxiety 2020; 37:146-155. [PMID: 31730745 DOI: 10.1002/da.22974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/14/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Sex differences in psychiatric disorders are common and could involve sex steroids. Aromatase, the product of the CYP19A1 gene, is the key enzyme in the conversion of androgen to estrogen. Whether CYP19A1 variants could be associated with depression differently in men and women has not been examined. METHODS This population-based study included 405 men and 602 women aged ≥65 years. A clinical level of depression (DEP) was defined as having a score ≥16 on the Center for Epidemiology Studies Depression scale or a diagnosis of current major depression based on the Mini-International Neuropsychiatric Interview and according to DSM-IV criteria. Seven single-nucleotide polymorphisms (SNPs) spanning the CYP19A1 gene were genotyped and circulating levels of estradiol and testosterone were determined. Multivariable analyses were adjusted for age, body mass index, ischemic pathologies, cognitive impairment, and anxiety. RESULTS Five SNPs were associated with DEP in women specifically and this varied according to a history of major depression (p-values .01 to .0005). Three SNPs were associated with an increased risk of late-life DEP in women without a history of major depression, while two SNPs were associated with a decreased DEP risk in women with a history of major depression and were also associated with higher estradiol levels. CONCLUSIONS Variants of the CYP19A1 gene appear to be susceptibility factors for late-life depression in a sex-specific manner. The polymorphisms decreasing the risk of recurrent depression in postmenopausal women also influence estradiol levels.
Collapse
Affiliation(s)
- Marie-Laure Ancelin
- Inserm, Neuropsychiatry: Epidemiological and Clinical Research, University of Montpellier, Montpellier, France
| | - Joanna Norton
- Inserm, Neuropsychiatry: Epidemiological and Clinical Research, University of Montpellier, Montpellier, France
| | - Marianne Canonico
- Centre for Research Epidemiology and Population Health, UVSQ, Inserm, Paris-Saclay University, Paris-South University, Villejuif, France
| | - Pierre-Yves Scarabin
- Centre for Research Epidemiology and Population Health, UVSQ, Inserm, Paris-Saclay University, Paris-South University, Villejuif, France
| | - Karen Ritchie
- Inserm, Neuropsychiatry: Epidemiological and Clinical Research, University of Montpellier, Montpellier, France.,Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanne Ryan
- Inserm, Neuropsychiatry: Epidemiological and Clinical Research, University of Montpellier, Montpellier, France.,Biological Neuropsychiatry and Dementia Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Abstract
Estrogens are critical in driving sex-typical social behaviours that are ethologically relevant in mammals. This is due to both production of local estrogens and signaling by these ligands, particularly in an interconnected set of nuclei called the social behavioural network (SBN). The SBN is a sexually dimorphic network studied predominantly in rodents that is thought to underlie the display of social behaviour in mammals. Signalling by the predominant endogenous estrogen, 17β-estradiol, can be either via the classical genomic or non-classical rapid pathway. In the classical genomic pathway, 17β-estradiol binds the intracellular estrogen receptors (ER) α and β which act as ligand-dependent transcription factors to regulate transcription. In the non-genomic pathway, 17β-estradiol binds a putative plasma membrane ER (mER) such as GPR30/GPER1 to rapidly signal via kinases or calcium flux. Though GPER1's role in sexual dimorphism has been explored to a greater extent in cardiovascular physiology, less is known about its role in the brain. In the last decade, activation of GPER1 has been shown to be important for lordosis and social cognition in females. In this review we will focus on several mechanisms that may contribute to sexually dimorphic behaviors including the colocalization of these estrogen receptors in the SBN, interplay between the signaling pathways activated by these different estrogen receptors, and the role of these receptors in development and the maintenance of the SBN, all of which remain underexplored.
Collapse
|
33
|
Lainez NM, Coss D. Obesity, Neuroinflammation, and Reproductive Function. Endocrinology 2019; 160:2719-2736. [PMID: 31513269 PMCID: PMC6806266 DOI: 10.1210/en.2019-00487] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022]
Abstract
The increasing occurrence of obesity has become a significant public health concern. Individuals with obesity have higher prevalence of heart disease, stroke, osteoarthritis, diabetes, and reproductive disorders. Reproductive problems include menstrual irregularities, pregnancy complications, and infertility due to anovulation, in women, and lower testosterone and diminished sperm count, in men. In particular, women with obesity have reduced levels of both gonadotropin hormones, and, in obese men, lower testosterone is accompanied by diminished LH. Taken together, these findings indicate central dysregulation of the hypothalamic-pituitary-gonadal axis, specifically at the level of the GnRH neuron function, which is the final brain output for the regulation of reproduction. Obesity is a state of hyperinsulinemia, hyperlipidemia, hyperleptinemia, and chronic inflammation. Herein, we review recent advances in our understanding of how these metabolic and immune changes affect hypothalamic function and regulation of GnRH neurons. In the latter part, we focus on neuroinflammation as a major consequence of obesity and discuss findings that reveal that GnRH neurons are uniquely positioned to respond to inflammatory changes.
Collapse
Affiliation(s)
- Nancy M Lainez
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
| | - Djurdjica Coss
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California
- Correspondence: Djurdjica Coss, PhD, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, 303 SOM Research Building, 900 University Avenue, Riverside, California 92521. E-mail:
| |
Collapse
|
34
|
Azcoitia I, Barreto GE, Garcia-Segura LM. Molecular mechanisms and cellular events involved in the neuroprotective actions of estradiol. Analysis of sex differences. Front Neuroendocrinol 2019; 55:100787. [PMID: 31513774 DOI: 10.1016/j.yfrne.2019.100787] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/27/2019] [Accepted: 09/07/2019] [Indexed: 12/12/2022]
Abstract
Estradiol, either from peripheral or central origin, activates multiple molecular neuroprotective and neuroreparative responses that, being mediated by estrogen receptors or by estrogen receptor independent mechanisms, are initiated at the membrane, the cytoplasm or the cell nucleus of neural cells. Estrogen-dependent signaling regulates a variety of cellular events, such as intracellular Ca2+ levels, mitochondrial respiratory capacity, ATP production, mitochondrial membrane potential, autophagy and apoptosis. In turn, these molecular and cellular actions of estradiol are integrated by neurons and non-neuronal cells to generate different tissue protective responses, decreasing blood-brain barrier permeability, oxidative stress, neuroinflammation and excitotoxicity and promoting synaptic plasticity, axonal growth, neurogenesis, remyelination and neuroregeneration. Recent findings indicate that the neuroprotective and neuroreparative actions of estradiol are different in males and females and further research is necessary to fully elucidate the causes for this sex difference.
Collapse
Affiliation(s)
- Iñigo Azcoitia
- Department of Cell Biology, Faculty of Biology, Universidad Complutense de Madrid, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - George E Barreto
- Department of Biological Sciences, School of Natural Sciences, University of Limerick, Limerick, Ireland.
| | - Luis M Garcia-Segura
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludables (CIBERFES), Instituto de Salud Carlos III, Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain; Instituto Cajal, CSIC, Avenida Doctor Arce 37, 28002 Madrid, Spain.
| |
Collapse
|
35
|
Brandt N, Rune GM. Sex-dependency of oestrogen-induced structural synaptic plasticity: Inhibition of aromatase versus application of estradiol in rodents. Eur J Neurosci 2019; 52:2548-2559. [PMID: 31403726 DOI: 10.1111/ejn.14541] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/19/2019] [Accepted: 08/01/2019] [Indexed: 12/21/2022]
Abstract
Sex-dependent differences in learning and memory formation in humans have been frequently shown. The mechanisms underlying the formation and retention of memories are assumed to involve synaptic plasticity in the hippocampus. Estradiol was shown to effect synaptic plasticity in the hippocampus of rodents. The effects after exogenous application of estradiol to animals frequently produce inconsistent results, in particular, if sex is not considered in the studies. Recently we provided evidence that locally synthesized estradiol plays an essential role on synaptic connectivity in the hippocampus of females but not of male mice. In females, inhibition of local estradiol synthesis leads to synapse loss, which results from impairment of long-term potentiation and dephosphorylation of cofilin, and thereby the destabilization of postsynaptic dendritic spines. This sex-dependency was also seen in the classical aromatase knock-out mouse. Intriguingly, no differences between sexes have been found in a conditional forebrain-specific aromatase knock-out mouse. Altogether, the findings underscore the necessity of including 'Sex as a Biological Variable' in studies of sex steroid-induced synaptic plasticity.
Collapse
Affiliation(s)
- Nicola Brandt
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|