1
|
Streifer M, Hilz EN, Raval R, Wylie DC, Gore AC. Transcriptomic analysis of effects of developmental PCB exposure in the hypothalamus of female rats. Mol Cell Endocrinol 2025; 599:112460. [PMID: 39798907 DOI: 10.1016/j.mce.2025.112460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
This study investigated the consequences of perinatal exposure to Aroclor 1221 (A1221), a weakly estrogenic polychlorinated biphenyl (PCB) mixture and known endocrine-disrupting chemical (EDC), in female rats. Previous work has shown behavioral and physiological effects of A1221, and the current study extended this work to comprehensive transcriptomic profiling of two hypothalamic regions involved in the control of reproduction: the arcuate nucleus (ARC) and anteroventral periventricular nucleus (AVPV). Female Sprague-Dawley rats were fed a cookie treated with a small volume of A1221 (1 mg/kg) or vehicle (3% DMSO in sesame oil) during pregnancy from gestational days 8-18 and after birth from postnatal (P) days 1-21, exposing the offspring via placental and lactational transfer. In female offspring, developmental, physiological, and hormonal effects of A1221 were relatively modest. However, because prior work has implicated this exposure in neurobehavioral disruptions, we sought to determine whether developmental programming of the brain transcriptome could underlie these latter phenotypes. We used 3' targeted RNA sequencing in the hypothalamus (arcuate nucleus, anteroventral periventricular nucleus) of experimental females at P8, 30, and 60 and identified significant alterations in gene expression and gene ontology (GO) terms in an age- and tissue-specific manner. Most notably, terms related to synaptic signaling, neurotransmitter regulation, immune response, and cellular structure were identified. Changes in pathways associated with synaptic functions and cellular metabolism were further identified, indicating that A1221 exposure can impact neurodevelopmental and neuroendocrine processes at a molecular level, even in the absence of overt developmental changes. These findings of molecular reprogramming may explain the behavioral effects of A1221 and highlight novel molecular targets and pathways that warrant further investigation to understand the effects of EDCs on the developing brain.
Collapse
Affiliation(s)
- Madeline Streifer
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA; Center for Molecular Carcinogenesis & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Emily N Hilz
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA; Center for Molecular Carcinogenesis & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Raj Raval
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Dennis C Wylie
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrea C Gore
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA; Center for Molecular Carcinogenesis & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
2
|
Padilla-Valdez MM, Santana-Bejarano MB, Godínez-Rubí M, Ortuño-Sahagún D, Rojas-Mayorquín AE. Prenatal Alcohol Exposure Disrupts CXCL16 Expression in Rat Hippocampus: Temporal and Sex Differences. Int J Mol Sci 2025; 26:1920. [PMID: 40076549 PMCID: PMC11900973 DOI: 10.3390/ijms26051920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Prenatal alcohol exposure (PAE) affects around 40,000 newborns every year and poses a significant health risk. Although much is already known about the neurotoxic mechanisms of PAE, new findings continue to emerge. Studies with mouse models show that PAE leads to overexpression of proinflammatory cytokines and chemokines in the brain, which disrupts important neurodevelopmental processes such as cell migration, survival and proliferation of neurons. The chemokine CXCL16 is overexpressed in the brain following various impairments, including PAE. This study shows that CXCL16 expression varies by developmental stage and sex, consistent with known sexual dimorphism in immune responses. In females, CXCL16 expression may be influenced by estrogen-related mechanisms, possibly related to the alcohol-mediated rebound effect described here. In contrast, the male hippocampus shows greater resilience to PAE-induced CXCL16 changes. Furthermore, the presence of CXCL16 in neuronal nuclei suggests a role in gene regulation, similar to other chemokines such as CCL5 and CXCL4. These findings shed light on the role of chemokines in hippocampal neuroplasticity and may pave the way for better treatment of fetal alcohol spectrum disorder (FASD).
Collapse
Affiliation(s)
- Mayra Madeleine Padilla-Valdez
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico;
| | - Margarita Belem Santana-Bejarano
- Laboratorio de Patología Diagnóstica e Inmunohistoquímica, Centro de Investigación y Diagnóstico en Patología, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico; (M.B.S.-B.); (M.G.-R.)
| | - Marisol Godínez-Rubí
- Laboratorio de Patología Diagnóstica e Inmunohistoquímica, Centro de Investigación y Diagnóstico en Patología, Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico; (M.B.S.-B.); (M.G.-R.)
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico;
| | | |
Collapse
|
3
|
Wang L, Li Z, Song Y, Li N, Liu XH, Wang D. Divergent Photoperiodic Responses in Hypothalamic Dio3 Expression and Gonadal Activity Between Offspring and Paternal Brandt's Voles. Animals (Basel) 2025; 15:469. [PMID: 40002951 PMCID: PMC11851783 DOI: 10.3390/ani15040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The postnatal development of gonadal glands in seasonal breeders, particularly small rodent species, is influenced by photoperiodic patterns. However, little research has been conducted on the effects of pattern similarity and age differentiation especially in molecular features. This study compares the postnatal development of gonadal glands and the expression of hypothalamic genes related to reproductive regulation in male offspring of Brandt's voles (Lasiopodomys brandtii) born under three types of changing photoperiodic patterns: increasing long photoperiod (ILP, 12 h + 3 min/day), natural increasing long photoperiods (NLPs), and decreasing short photoperiods (DSPs, 12 h - 3 min/day), as well as in their paternal voles exposed to these patterns at the same period. Results indicate that over the course of 12 postnatal weeks, gonadal development, including organ masses and serum testosterone levels, exhibited similar profiles between the ILP and NLP groups, which were significantly higher than those observed in DSP offspring. Hypothalamic type 3 iodothyronine deiodinase (Dio3) exhibited significantly higher expression in the DSP group from postnatal week 4 to 8 compared to the other two groups. These physiological and molecular differences gradually decreased with age in offspring, but were never observed in the paternal voles, indicating divergent photoperiodic responses between the two ages. The synchronous profiles observed between hypothalamic Dio3 expression and gonadal activities underscore its crucial role in interpreting photoperiodic signals and regulating gonadal development in Brandt's voles.
Collapse
Affiliation(s)
- Lewen Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
| | - Zhengguang Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
| | - Ying Song
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
| | - Ning Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
| | - Xiao-Hui Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
| | - Dawei Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (L.W.); (Z.L.); (Y.S.); (X.-H.L.)
- Western Agricultural Research Center, Chinese Academy of Agricultural Sciences, Changji 831100, China
- Key Laboratory of Biohazard Monitoring and Green Prevention and Control in Artificial Grassland, Ministry of Agriculture and Rural Affairs, Institute of Grassland Research, Chinese Academy of Agricultural Sciences, Hohhot 010010, China
| |
Collapse
|
4
|
Forrester-Fronstin Z, Barrett AR, Mondschein AS, Johnson JM, Cordes CN, Lawton-Stone TS, Schatz KC, Paul MJ. Exogenous estradiol impacts anxiety-like behavior of juvenile male and female Siberian hamsters in a dose-dependent manner. Horm Behav 2025; 167:105674. [PMID: 39731972 DOI: 10.1016/j.yhbeh.2024.105674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Anxiety is among the most prevalent mental health issues in children. While it is well established that gonadal steroids influence anxiety-like behavior in adulthood, a potential role in prepubertal juveniles has been overlooked because it is commonly thought that the gonads are quiescent during the juvenile period. However, the juvenile gonads secrete measurable amounts of steroids, and we have recently found that prepubertal ovariectomy decreases anxiety-like behavior of juvenile Siberian hamsters in the light/dark box test. The present study tested whether an injection of estradiol benzoate (1 μg or 10 μg, SC) to gonadectomized hamsters (Exp. 1) or chronic suppression of endogenous estradiol with the aromatase inhibitor, letrozole (2 mg/kg, PO), to intact hamsters (Exp. 2) affects anxiety-like behavior in the light/dark box test during the juvenile phase. Estradiol benzoate altered anxiety-like behavior of both male and female juveniles in a dose-dependent manner, with anxiolytic actions at the low dose, but no effect at the high dose. Similar effects were seen for activity measures, albeit only in females. Letrozole suppressed uterine weights demonstrating an active role for endogenous estradiol during the juvenile phase. Anxiety-like behavior, however, was impacted by the administration procedure itself, preventing conclusions on letrozole's actions on behavior. While the role for endogenous estradiol in juvenile anxiety-like behavior remains unresolved, the present findings indicate that the neural centers regulating affective behavior are responsive to exogenous estradiol prior to puberty. These findings highlight the potential impact of exogenous estrogen exposures on juvenile affective behavior.
Collapse
Affiliation(s)
| | - Abigal R Barrett
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | | | - Jordan M Johnson
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Chloe N Cordes
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | | | - Kelcie C Schatz
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA
| | - Matthew J Paul
- Department of Psychology, University at Buffalo, SUNY, Buffalo, NY, USA; Neuroscience Program, University at Buffalo, SUNY, Buffalo, NY, USA; Evolution, Ecology, and Behavior Program, University at Buffalo, SUNY, NY, USA
| |
Collapse
|
5
|
Vazakidou P, Bouftas N, Heinzelmann M, Johansson HKL, Svingen T, Leonards PEG, van Duursen MBM. Minor changes to circulating steroid hormones in female rats after perinatal exposure to diethylstilbestrol or ketoconazole. Reprod Toxicol 2024; 130:108726. [PMID: 39326550 DOI: 10.1016/j.reprotox.2024.108726] [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: 07/18/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Current chemical test strategies lack sensitive markers for detecting female reproductive toxicity caused by endocrine disrupting chemicals (EDCs). In search of a potentially sensitive readout, the steroidogenic disrupting effects of the well-known EDCs ketoconazole (KTZ) and diethylstilbestrol (DES) were investigated in vitro and on circulating steroid hormones in perinatally exposed female Sprague-Dawley rats. Twenty-one steroid hormones were analysed using LC-MS/MS in plasma from female rat offspring at postnatal day (PD) 6, 14, 22, 42 and 90. Most circulating steroid hormone levels increased with age except for estrone (E1), estradiol (E2) and backdoor pathway androsterone (ANDROST), which decreased after PD 22. Perinatal exposure to DES did not affect circulating steroid hormone levels at any dose or age compared to controls. KTZ exposure resulted in dose-dependent increase of corticosterone (CORTICO) at PD 6 and PD 14, with statistical significance only at PD 14. In the in vitro gold standard H295R steroidogenesis assay, twenty-one steroid hormones were measured instead of only T and E2. DES had subtle effects on steroidogenesis, whereas KTZ decreased most steroid hormones, but increased CORTICO, progesterone (P4), estriol (E3) initially (around 0.1-1 µM) before decreasing. Our data suggests that circulating steroidomic profiling may not be a sensitive readout for EDC-induced female reproductive toxicity. Further studies are needed to associate H295R assay steroidomic profiles with in vivo profiles, especially in target tissues such as adrenals or gonads. Expanding the H295R steroidogenic assay to include a comprehensive steroidomic profile may enhance its regulatory applicability.
Collapse
Affiliation(s)
- Paraskevi Vazakidou
- Environment and Health, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands
| | - Nora Bouftas
- Environment and Health, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands
| | - Manuel Heinzelmann
- Environment and Health, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands
| | - Hanna K L Johansson
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | - Pim E G Leonards
- Environment and Health, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands
| | - Majorie B M van Duursen
- Environment and Health, Amsterdam Institute for Life and Environment, Vrije Universiteit Amsterdam, De Boelelaan 1085, Amsterdam 1081 HV, the Netherlands.
| |
Collapse
|
6
|
Streifer M, Thompson LM, Mendez SA, Gore AC. Neuroendocrine and Developmental Impacts of Early Life Exposure to EDCs. J Endocr Soc 2024; 9:bvae195. [PMID: 39659541 PMCID: PMC11631349 DOI: 10.1210/jendso/bvae195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Indexed: 12/12/2024] Open
Abstract
Polychlorinated biphenyls (PCBs) pose a global challenge to environmental and human health. Although toxic and carcinogenic at higher exposure levels, at lower concentrations they can act as endocrine-disrupting chemicals. Individuals are more vulnerable to endocrine-disrupting effects of PCB exposures during the perinatal period, when the neuroendocrine system is developing, although assessing the full impact of PCB exposure is difficult because of the often-latent onset of adverse effects. The goal of this study was to determine developmental effects of an estrogenic PCB mixture, Aroclor 1221 (A1221), on KNDy and kisspeptin neuron numbers in the hypothalamic arcuate nucleus and anteroventral periventricular nucleus (AVPV), together with measures of hypothalamic-pituitary-gonadal hormones and postnatal development. We conducted RNAscope of kisspeptin, prodynorphin, neurokinin B, and estrogen receptor alpha genes in the P30 hypothalamus. Early-life PCBs caused small but significant changes in development (body weight and anogenital index) but had no effect on puberty. We found sex-specific effects of treatment on serum LH, FSH, and estradiol in a sex- and developmental age-dependent manner. RNAscope results revealed increased prodynorphin in the AVPV of male rats, but no effects on kisspeptin or neurokinin B in AVPV or arcuate nucleus. An unexpected species difference was found: we were unable to detect prodynorphin coexpression with kisspeptin within KNDy neurons in rats, unlike mice, sheep, and primates. These data show that early-life PCBs can induce developmental and hormonal changes that together with other reports showing latent effects on behavior and the hypothalamic-pituitary-gonadal axis, indicate adverse endocrine and neurobehavioral outcomes.
Collapse
Affiliation(s)
- Madeline Streifer
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lindsay M Thompson
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Skylar A Mendez
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea C Gore
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
7
|
Hsieh KL, Sun TB, Huang KH, Lin CH, Tang LY, Liu CL, Chao CM, Chang CP. Hyperbaric oxygen preconditioning normalizes scrotal temperature, sperm quality, testicular structure, and erectile function in adult male rats subjected to exertional heat injury. Mol Cell Endocrinol 2024; 584:112175. [PMID: 38341020 DOI: 10.1016/j.mce.2024.112175] [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: 11/28/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Testicular hyperthermia has been noted in men who work in high ambient temperatures. Scrotal temperatures above the normal range caused germ cell loss in the testes and resulted in male subfertility. In adult male rats, exercising at a higher environmental temperature (36 °C with relative humidity of 50%, 52 min) caused exertional heat stroke (EHS) characterized by scrotal hyperthermia, impaired sperm quality, dysmorphology in testes, prostates and bladders, and erectile dysfunction. Here, we aim to ascertain whether hyperbaric oxygen preconditioning (HBOP: 100% O2 at 2.0 atm absolute [ATA] for 2 h daily for 14 days consequently before the onset of EHS) is able to prevent the problem of EHS-induced sterility, testes, prostates, and bladders dysmorphology and erectile dysfunction. At the end of exertional heat stress compared to normobaric air (NBA or non-HBOP) rats, the HBOP rats exhibited lower body core temperature (40 °C vs. 43 °C), lower scrotal temperature (34 °C vs. 36 °C), lower neurological severity scores (2.8 vs. 5.8), higher erectile ability, (5984 mmHg-sec vs. 3788 mmHg-sec), higher plasma testosterone (6.8 ng/mL vs. 3.5 ng/mL), lower plasma follicle stimulating hormone (196.3 mIU/mL vs. 513.8 mIU/mL), lower plasma luteinizing hormone (131 IU/L vs. 189 IU/L), lower plasma adrenocorticotropic hormone (5136 pg/mL vs. 6129 pg/mL), lower plasma corticosterone (0.56 ng/mL vs. 1.18 ng/mL), lower sperm loss and lower values of histopathological scores for epididymis, testis, seminal vesicle, prostate, and bladder. Our data suggest that HBOP reduces body core and scrotal hyperthermia and improves sperm loss, testis/prostate/bladder dysmorphology, and erectile dysfunction after EHS in rats.
Collapse
Affiliation(s)
- Kun-Lin Hsieh
- Division of Urology, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan; Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Tzong-Bor Sun
- Department of Hyperbaric Oxygen Medicine, Chi-Mei Medical Center, Tainan, Taiwan; Division of Plastic Surgery, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan.
| | - Kuan-Hua Huang
- Division of Urology, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan.
| | - Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan; Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.
| | - Ling-Yu Tang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.
| | - Chien-Liang Liu
- Division of Urology, Department of Surgery, Chi-Mei Medical Center, Tainan, Taiwan.
| | - Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan; Department of Dental Laboratory Technology, Min-Hwei College of Health Care Management, Tainan, Taiwan.
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan.
| |
Collapse
|
8
|
Oliveira GDF, Nguyen AT, Carreras-Simons L, Niepsuj T, Gadelhak SH, Johnson AK, Abdalla A, Lev E, Torres Román SG, Fuchs SN, Jorgensen JS, Farhat WA, Auger AP. Puberty Blocker, Leuprolide, Reduces Sex Differences in Rough-and-Tumble Play and Anxiety-like Behavior in Juvenile Rats. Endocrinology 2024; 165:bqae046. [PMID: 38597659 PMCID: PMC11033216 DOI: 10.1210/endocr/bqae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
We examined the effect of the puberty blocker, leuprolide acetate, on sex differences in juvenile rough-and-tumble play behavior and anxiety-like behavior in adolescent male and female rats. We also evaluated leuprolide treatment on gonadal and pituitary hormone levels and activity-regulated cytoskeleton-protein messenger RNA levels within the adolescent amygdala, a region important both for rough-and-tumble play and anxiety-like behavior. Our findings suggest that leuprolide treatment lowered anxiety-like behavior during adolescent development, suggesting that the maturation of gonadotropin-releasing hormone systems may be linked to increased anxiety. These data provide a potential new model to understand the emergence of increased anxiety triggered around puberty. Leuprolide also reduced masculinized levels of rough-and-tumble play behavior, lowered follicle-stimulating hormone, and produced a consistent pattern of reducing or halting sex differences of hormone levels, including testosterone, growth hormone, thyrotropin, and corticosterone levels. Therefore, leuprolide treatment not only pauses sexual development of peripheral tissues, but also reduces sex differences in hormones, brain, and behavior, allowing for better harmonization of these systems following gender-affirming hormone treatment. These data contribute to the intended use of puberty blockers in stopping sex differences from developing further with the potential benefit of lowering anxiety-like behavior.
Collapse
Affiliation(s)
- Gabriela de Faria Oliveira
- Wisconsin National Primate Research Center, Madison, WI 53715, USA
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Amber T Nguyen
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | | | - Thomas Niepsuj
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Salma H Gadelhak
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Aimee K Johnson
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Ashwakh Abdalla
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Eden Lev
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Sofia G Torres Román
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Samantha N Fuchs
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Joan S Jorgensen
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI 53706, USA
| | - Walid A Farhat
- Division of Pediatric Urology, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI 53705, USA
| | - Anthony P Auger
- Department of Psychology, University of Wisconsin–Madison, Madison, WI 53706, USA
| |
Collapse
|
9
|
Mao S, Dong S, Hou B, Li Y, Sun B, Guo Y, Deng M, Liu D, Liu G. Transcriptome analysis reveals pituitary lncRNA, circRNA and mRNA affecting fertility in high- and low-yielding goats. Front Genet 2023; 14:1303031. [PMID: 38152654 PMCID: PMC10751935 DOI: 10.3389/fgene.2023.1303031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023] Open
Abstract
The pituitary gland serves as the central endocrine regulator of growth, reproduction, and metabolism and plays a crucial role in the reproductive process of female animals. Transcriptome analysis was conducted using pituitary gland samples from Leizhou goats with varying levels of fecundity to investigate the effects of long noncoding RNA (lncRNA), circular RNA (circRNA), and mRNA regulation on pituitary hormone secretion and its association with goat fecundity. The analysis aimed to identify lncRNAs, circRNAs, and mRNAs that influence the fertility of Leizhou goats. GO and KEGG enrichment analyses were performed on differentially expressed lncRNAs, circRNAs, and mRNAs and revealed considerable enrichment in pathways, such as regulation of hormone secretion, germ cell development, and gonadotropin-releasing hormone secretion. The pituitary lncRNAs (ENSCHIT00000010293, ENSCHIT00000010304, ENSCHIT00000010306, ENSCHIT00000010290, ENSCHIT00000010298, ENSCHIT00000006769, ENSCHIT00000006767, ENSCHIT00000006921, and ENSCHIT00000001330) and circRNAs (chicirc_029285, chicirc_026618, chicirc_129655, chicirc_018248, chicirc_122554, chicirc_087101, and chicirc_078945) identified as differentially expressed regulated hormone secretion in the pituitary through their respective host genes. Additionally, differential mRNAs (GABBR2, SYCP1, HNF4A, CBLN1, and CDKN1A) influenced goat fecundity by affecting hormone secretion in the pituitary gland. These findings contribute to the understanding of the molecular mechanisms underlying pituitary regulation of fecundity in Leizhou goats.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dewu Liu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Guangbin Liu
- College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
10
|
Abeledo-Machado A, Peña-Zanoni M, Bornancini D, Díaz-Torga G. Revealing Sexual Dimorphism in Prolactin Regulation From Early Postnatal Development to Adulthood in Murine Models. J Endocr Soc 2023; 8:bvad146. [PMID: 38045876 PMCID: PMC10690727 DOI: 10.1210/jendso/bvad146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Indexed: 12/05/2023] Open
Abstract
Serum prolactin (PRL) levels exhibit a gradual rise both in male and female rats from birth to adulthood, with females consistently displaying higher levels compared to age-matched males. This pattern has traditionally been attributed to the development and maturation of endocrine and neuroendocrine networks responsible for regulating PRL synthesis and secretion. However, the effect of dopamine (DA), which acts as an inhibitory factor on lactotroph function, also increases from birth to puberty, particularly in females. Nonetheless, the secretion of PRL remains higher in females compared to males. On the other hand, the observed sex differences in serum PRL levels during early postnatal development cannot be attributed to the influence of estradiol (E2). While serum E2 levels gradually increase after birth, only after 45 days of life do the disparities in E2 levels between females and males become evident. These observations collectively suggest that neither the maturation of hypothalamic DA regulation nor the rise in E2 levels can account for the progressive and sustained elevation in serum PRL levels and the observed sexual dimorphism during postnatal development. This review highlights the importance of recent discoveries in animal models that shed light on inhibitory mechanisms in the control of PRL secretion within the pituitary gland itself, that is intrapituitary mechanisms, with a specific emphasis on the role of transforming growth factor β1 and activins in PRL secretion.
Collapse
Affiliation(s)
- Alejandra Abeledo-Machado
- Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, CONICET, Buenos Aires 1428, Argentina
| | - Milagros Peña-Zanoni
- Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, CONICET, Buenos Aires 1428, Argentina
| | - Dana Bornancini
- Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, CONICET, Buenos Aires 1428, Argentina
| | - Graciela Díaz-Torga
- Instituto de Biología y Medicina Experimental (IBYME), Fundación IBYME, CONICET, Buenos Aires 1428, Argentina
| |
Collapse
|
11
|
Yamada K, Nagae M, Mano T, Tsuchida H, Hazim S, Goto T, Sanbo M, Hirabayashi M, Inoue N, Uenoyama Y, Tsukamura H. Sex difference in developmental changes in visualized Kiss1 neurons in newly generated Kiss1-Cre rats. J Reprod Dev 2023; 69:227-238. [PMID: 37518187 PMCID: PMC10602768 DOI: 10.1262/jrd.2023-019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Hypothalamic kisspeptin neurons are master regulators of mammalian reproduction via direct stimulation of gonadotropin-releasing hormone and consequent gonadotropin release. Here, we generated novel Kiss1 (kisspeptin gene)-Cre rats and investigated the developmental changes and sex differences in visualized Kiss1 neurons of Kiss1-Cre-activated tdTomato reporter rats. First, we validated Kiss1-Cre rats by generating Kiss1-expressing cell-specific Kiss1 knockout (Kiss1-KpKO) rats, which were obtained by crossing the current Kiss1-Cre rats with Kiss1-floxed rats. The resulting male Kiss1-KpKO rats lacked Kiss1 expression in the brain and exhibited hypogonadotropic hypogonadism, similar to the hypogonadal phenotype of global Kiss1 KO rats. Histological analysis of Kiss1 neurons in Kiss1-Cre-activated tdTomato reporter rats revealed that tdTomato signals in the anteroventral periventricular nucleus (AVPV) and arcuate nucleus (ARC) were not affected by estrogen, and that tdTomato signals in the ARC, AVPV, and medial amygdala (MeA) were sexually dimorphic. Notably, neonatal AVPV tdTomato signals were detected only in males, but a larger number of tdTomato-expressing cells were detected in the AVPV and ARC, and a smaller number of cells in the MeA was detected in females than in males at postpuberty. These findings suggest that Kiss1-visualized rats can be used to examine the effect of estrogen feedback mechanisms on Kiss1 expression in the AVPV and ARC. Moreover, the Kiss1-Cre and Kiss1-visualized rats could be valuable tools for further detailed analyses of sexual differentiation in the brain and the physiological role of kisspeptin neurons across the brain in rats.
Collapse
Affiliation(s)
- Koki Yamada
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Mayuko Nagae
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Tetsuya Mano
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Hitomi Tsuchida
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Safiullah Hazim
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Teppei Goto
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Makoto Sanbo
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Masumi Hirabayashi
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, Aichi 444-8787, Japan
| | - Naoko Inoue
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Yoshihisa Uenoyama
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| | - Hiroko Tsukamura
- Laboratory of Animal Reproduction, Graduate School of Bioagricultural Sciences, Nagoya University, Aichi 464-8601, Japan
| |
Collapse
|
12
|
Frau R, Melis M. Sex-specific susceptibility to psychotic-like states provoked by prenatal THC exposure: Reversal by pregnenolone. J Neuroendocrinol 2023; 35:e13240. [PMID: 36810840 DOI: 10.1111/jne.13240] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023]
Abstract
Sociocultural attitudes towards cannabis legalization contribute to the common misconception that it is a relatively safe drug and its use during pregnancy poses no risk to the fetus. However, longitudinal studies demonstrate that maternal cannabis exposure results in adverse outcomes in the offspring, with a heightened risk for developing psychopathology. One of the most reported psychiatric outcomes is the proneness to psychotic-like experiences during childhood. How exposure to cannabis during gestation increases psychosis susceptibility in children and adolescents remains elusive. Preclinical research has indicated that in utero exposure to the major psychoactive component of cannabis, delta-9-tetrahydrocannabinol (THC), deranges brain developmental trajectories towards vulnerable psychotic-like endophenotypes later in life. Here, we present how prenatal THC exposure (PCE) deregulates mesolimbic dopamine development predisposing the offspring to schizophrenia-relevant phenotypes, exclusively when exposed to environmental challenges, such as stress or THC. Detrimental effects of PCE are sex-specific because female offspring do not display psychotic-like outcomes upon exposure to these challenges. Moreover, we present how pregnenolone, a neurosteroid that showed beneficial properties on the effects elicited by cannabis intoxication, normalizes mesolimbic dopamine function and rescues psychotic-like phenotypes. We, therefore, suggest this neurosteroid as a safe "disease-modifying" aid to prevent the onset of psychoses in vulnerable individuals. Our findings corroborate clinical evidence and highlight the relevance of early diagnostic screening and preventative strategies for young individuals at risk for mental diseases, such as male PCE offspring.
Collapse
Affiliation(s)
- Roberto Frau
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
- The Guy Everett Laboratory for Neuroscience, University of Cagliari, Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Monserrato, Italy
| |
Collapse
|
13
|
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
|
14
|
Methods and Challenges in Investigating Sex-Specific Consequences of Social Stressors in Adolescence in Rats: Is It the Stress or the Social or the Stage of Development? Curr Top Behav Neurosci 2021; 54:23-58. [PMID: 34455576 DOI: 10.1007/7854_2021_245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Adolescence is a time of social learning and social restructuring that is accompanied by changes in both the hypothalamic-pituitary-gonadal axis and the hypothalamic-pituitary-adrenal (HPA) axis. The activation of these axes by puberty and stressors, respectively, shapes adolescent development. Models of social stress in rats are used to understand the consequences of perturbations of the social environment for ongoing brain development. This paper reviews the challenges in investigating the sex-specific consequences of social stressors, sex differences in the models of social stress used in rats and the sex-specific effects on behaviour and provides an overview of sex differences in HPA responding to stressors, the variability in pubertal development and in strains of rats that require consideration in conducting such research, and directions for future research.
Collapse
|
15
|
Al Abed AS, Reynolds NJ, Dehorter N. A Second Wave for the Neurokinin Tac2 Pathway in Brain Research. Biol Psychiatry 2021; 90:156-164. [PMID: 33867115 DOI: 10.1016/j.biopsych.2021.02.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/19/2022]
Abstract
Despite promising advances in basic research of the neurokinin B/Tac2 pathway in both animals and humans, clinical applications are yet to be implemented. This is likely because of our limited understanding of the action of the pathway in the brain. While this system controls neuronal activity in multiple regions, the precise impact of Tac2-induced cellular responses on behavior remains unclear. Recently, elegant studies revealed a key contribution to stress-related behaviors and memory. Here, we discuss the crucial importance of bridging the gap between the Tac2 pathway's involvement in cell physiology and cognition to comprehend its role in health and disease. We propose that a better understanding of the Tac2 pathway in the brain could provide an essential perspective for basic investigations, which in turn will feed clinical research.
Collapse
Affiliation(s)
- A Shaam Al Abed
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Nathan J Reynolds
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Nathalie Dehorter
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
16
|
Srancikova A, Bacova Z, Bakos J. The epigenetic regulation of synaptic genes contributes to the etiology of autism. Rev Neurosci 2021; 32:791-802. [PMID: 33939901 DOI: 10.1515/revneuro-2021-0014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022]
Abstract
Epigenetic mechanisms greatly affect the developing brain, as well as the maturation of synapses with pervasive, long-lasting consequences on behavior in adults. Substantial evidence exists that implicates dysregulation of epigenetic mechanisms in the etiology of neurodevelopmental disorders. Therefore, this review explains the role of enzymes involved in DNA methylation and demethylation in neurodevelopment by emphasizing changes of synaptic genes and proteins. Epigenetic causes of sex-dependent differences in the brain are analyzed in conjunction with the pathophysiology of autism spectrum disorders. Special attention is devoted to the epigenetic regulation of the melanoma-associated antigen-like gene 2 (MAGEL2) found in Prader-Willi syndrome, which is known to be accompanied by autistic symptoms.
Collapse
Affiliation(s)
- Annamaria Srancikova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Zuzana Bacova
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
| | - Jan Bakos
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| |
Collapse
|
17
|
Hernandez Scudder ME, Young RL, Thompson LM, Kore P, Crews D, Hofmann HA, Gore AC. EDCs Reorganize Brain-Behavior Phenotypic Relationships in Rats. J Endocr Soc 2021; 5:bvab021. [PMID: 33928200 PMCID: PMC8055178 DOI: 10.1210/jendso/bvab021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
All species, including humans, are exposed to endocrine-disrupting chemicals (EDCs). Previous experiments have shown behavioral deficits caused by EDCs that have implications for social competence and sexual selection. The neuromolecular mechanisms for these behavioral changes induced by EDCs have not been thoroughly explored. Here, we tested the hypothesis that EDCs administered to rats during a critical period of embryonic brain development would lead to the disruption of normal social preference behavior, and that this involves a network of underlying gene pathways in brain regions that regulate these behaviors. Rats were exposed prenatally to human-relevant concentrations of EDCs (polychlorinated biphenyls [PCBs], vinclozolin [VIN]), or vehicle. In adulthood, a sociosexual preference test was administered. We profiled gene expression of in preoptic area, medial amygdala, and ventromedial nucleus. Prenatal PCBs impaired sociosexual preference in both sexes, and VIN disrupted this behavior in males. Each brain region had unique sets of genes altered in a sex- and EDC-specific manner. The effects of EDCs on individual traits were typically small, but robust; EDC exposure changed the relationships between gene expression and behavior, a pattern we refer to as dis-integration and reconstitution. These findings underscore the effects that developmental exposure to EDCs can have on adult social behavior, highlight sex-specific and individual variation in responses, and provide a foundation for further work on the disruption of genes and behavior after prenatal exposure to EDCs.
Collapse
Affiliation(s)
| | - Rebecca L Young
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Lindsay M Thompson
- Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Pragati Kore
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - David Crews
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hans A Hofmann
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA.,Department of Integrative Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Andrea C Gore
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA.,Division of Pharmacology & Toxicology, The University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
18
|
Oemcke LA, Anderson RC, Rakonjac J, McNabb WC, Roy NC. Whole tissue homogenization preferable to mucosal scraping in determining the temporal profile of segmented filamentous bacteria in the ileum of weanling rats. Access Microbiol 2021; 3:000218. [PMID: 34151170 PMCID: PMC8209713 DOI: 10.1099/acmi.0.000218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/04/2021] [Indexed: 01/12/2023] Open
Abstract
Segmented filamentous bacteria (SFB) are thought to play a role in small intestine immunological maturation. Studies in weanling mice have shown a positive correlation between ileal SFB abundance and plasma and faecal interleukin 17 (IL-17) and immunoglobulin A (IgA) concentrations. Although the first observation of SFB presence was reported in rats, most studies use mice. The size of the mouse ileum is a limitation whereas the rat could be a suitable alternative for sufficient samples. Changes in SFB abundance over time in rats were hypothesized to follow the pattern reported in mice and infants. We characterized the profile of SFB colonization in the ileum tissue and contents and its correlation with two immune markers of gastrointestinal tract (GIT) maturation. We also compared two published ileum collection techniques to determine which yields data on SFB abundance with least variability. Whole ileal tissue and ileal mucosal scrapings were collected from 20- to 32-day-old Sprague-Dawley rats. SFB abundance was quantified from proximal, middle and distal ileal tissues, contents and faeces by quantitative PCR using SFB-specific primers. Antibody-specific ELISAs were used to determine IL-17 and IgA concentrations. Significant differences in SFB abundance were observed from whole and scraped tissues peaking at day 22. Variability in whole ileum data was less, favouring it as a better collection technique. A similar pattern of SFB abundance was observed in ileum contents and faeces peaking at day 24, suggesting faeces can be a proxy for ileal SFB abundance. SFB abundance at day 26 was higher in females than males across all samples. There were significant differences in IgA concentration between days 20, 30 and 32 and none in IL-17 concentration, which was different from reports in mice and infants.
Collapse
Affiliation(s)
- Linda A. Oemcke
- Riddet Institute, Massey University, Palmerston North, New Zealand
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand
- AgResearch, Grasslands Research Centre, Palmerston North, New Zealand
| | - Rachel C. Anderson
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch, Grasslands Research Centre, Palmerston North, New Zealand
| | - Jasna Rakonjac
- School of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| | - Warren C. McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C. Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
- Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
19
|
Annie L, Gurusubramanian G, Roy VK. Inhibition of visfatin/NAMPT affects ovarian proliferation, apoptosis, and steroidogenesis in pre-pubertal mice ovary. J Steroid Biochem Mol Biol 2020; 204:105763. [PMID: 32987128 DOI: 10.1016/j.jsbmb.2020.105763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 12/15/2022]
Abstract
Pubertal ovarian function might be dependent on the factors present in the pre-pubertal stages. Visfatin regulates ovarian steroidogenesis in adult. To date, no study has investigated the role of visfatin either in pre-pubertal or pubertal mice ovary. Thus, we investigated the role of visfatin in pre-pubertal mice ovary in relation to steroidogenesis and proliferation and apoptosis in vitro by inhibiting the endogenous visfatin by a specific inhibitor, FK866. Inhibition of visfatin increased the estrogen secretion and also up-regulated the expression of CYP11A1, 17βHSD and CYP19A1 in mice ovary. Furthermore, active caspase3 was up-regulated along with the down-regulation of BAX and BCL2 in the pre-pubertal ovary after visfatin inhibition. The expression of GCNA, PCNA, and BrdU labeling was also decreased by FK866 treatment. These results suggest that visfatin inhibits steroidogenesis, increases proliferation, and suppresses apoptosis in the pre-pubertal mice ovary. So, visfatin is a new regulator of ovary function in pre-pubertal mice.
Collapse
Affiliation(s)
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl, Mizoram 796 004, India.
| |
Collapse
|
20
|
Bizzozzero-Hiriart M, Di Giorgio NP, Libertun C, Lux-Lantos V. GABAergic input through GABA B receptors is necessary during a perinatal window to shape gene expression of factors critical to reproduction such as Kiss1. Am J Physiol Endocrinol Metab 2020; 318:E901-E919. [PMID: 32286880 DOI: 10.1152/ajpendo.00547.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Lack of GABAB receptors in GABAB1 knockout mice decreases neonatal ARC kisspeptin 1 (Kiss1) expression in the arcuate nucleus of the hypothalamus (ARC) in females, which show impaired reproduction as adults. Our aim was to selectively impair GABAB signaling during a short postnatal period to evaluate its impact on the reproductive system. Neonatal male and female mice were injected with the GABAB antagonist CGP 55845 (CGP, 1 mg/kg body wt sc) or saline from postnatal day 2 (PND2) to PND6, three times per day (8 AM, 1 PM, and 6 PM). One group was killed on PND6 for collection of blood samples (hormones by radioimmunoassay), brains for gene expression in the anteroventral periventricular nucleus-periventricular nucleus continuum (AVPV/PeN), and ARC micropunches [quantitative PCR (qPCR)] and gonads for qPCR, hormone contents, and histology. A second group of mice was injected with CGP (1 mg/kg body wt sc) or saline from PND2 to PND6, three times per day (8 AM, 1 PM, and 6 PM), and left to grow to adulthood. We measured body weight during development and parameters of sexual differentiation, puberty onset, and estrous cycles. Adult mice were killed, and trunk blood (hormones), brains for qPCR, and gonads for qPCR and hormone contents were obtained. Our most important findings on PND6 include the CGP-induced decrease in ARC Kiss1 and increase in neurokinin B (Tac2) in both sexes; the decrease in AVPV/PeN tyrosine hydroxylase (Th) only in females; the increase in gonad estradiol content in both sexes; and the increase in primordial follicles and decrease in primary and secondary follicles. Neonatally CGP-treated adults showed decreased ARC Kiss1 and ARC gonadotropin-releasing hormone (Gnrh1) and increased ARC glutamic acid decarboxylase 67 (Gad1) only in males; increased ARC GABAB receptor subunit 1 (Gabbr1) in both sexes; and decreased AVPV/PeN Th only in females. We demonstrate that ARC Kiss1 expression is chronically downregulated in males and that the normal sex difference in AVPV/PeN Th expression is abolished. In conclusion, neonatal GABAergic input through GABAB receptors shapes gene expression of factors critical to reproduction.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Arcuate Nucleus of Hypothalamus/drug effects
- Arcuate Nucleus of Hypothalamus/metabolism
- Estradiol/metabolism
- Female
- Follicle Stimulating Hormone/metabolism
- GABA-B Receptor Antagonists/pharmacology
- Gene Expression Regulation, Developmental/drug effects
- Gene Expression Regulation, Developmental/physiology
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/metabolism
- Gonadotropin-Releasing Hormone/genetics
- Gonadotropin-Releasing Hormone/metabolism
- Hypothalamus, Anterior/drug effects
- Hypothalamus, Anterior/metabolism
- Kisspeptins/genetics
- Kisspeptins/metabolism
- Luteinizing Hormone/metabolism
- Male
- Mice
- Ovary/drug effects
- Ovary/metabolism
- Phosphinic Acids/pharmacology
- Propanolamines/pharmacology
- Protein Precursors/genetics
- Protein Precursors/metabolism
- Puberty/drug effects
- Puberty/genetics
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, GABA-B/genetics
- Receptors, GABA-B/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Reproduction/drug effects
- Reproduction/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sex Differentiation/drug effects
- Sex Differentiation/genetics
- Tachykinins/genetics
- Tachykinins/metabolism
- Testis/drug effects
- Testis/metabolism
- Testosterone/metabolism
- Tyrosine 3-Monooxygenase/genetics
- Tyrosine 3-Monooxygenase/metabolism
Collapse
Affiliation(s)
- Marianne Bizzozzero-Hiriart
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Noelia P Di Giorgio
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Carlos Libertun
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Departamento de Fisiología, Facultad de Medicina, Universidad de Buenos Aires, Argentina
| | - Victoria Lux-Lantos
- Laboratorio de Neuroendocrinología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
21
|
Gegenhuber B, Tollkuhn J. Signatures of sex: Sex differences in gene expression in the vertebrate brain. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2020; 9:e348. [PMID: 31106965 PMCID: PMC6864223 DOI: 10.1002/wdev.348] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/10/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.
Collapse
Affiliation(s)
- Bruno Gegenhuber
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York
| | | |
Collapse
|
22
|
Kyne RF, Barrett AR, Brown LM, Paul MJ. Prepubertal ovarian inhibition of Light/Dark Box exploration and novel object investigation in juvenile Siberian hamsters. Horm Behav 2019; 115:104559. [PMID: 31310759 DOI: 10.1016/j.yhbeh.2019.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 11/24/2022]
Abstract
The overwhelming majority of research on the role of gonadal hormones in behavioral development has focused on perinatal, pubertal, or adult life stages. The juvenile period has been overlooked because it is thought to be a time of gonadal quiescence. In the present study, we tested whether prepubertal gonadectomy impacts the behavior of male and female juvenile hamsters on the Light/Dark Box, Novel Object, and Social Approach tests (Experiment 1) and compared these findings to those obtained after adult gonadectomy (Experiment 2). Prepubertal ovariectomy increased exploration (i.e. time spent in the light zone of the Light/Dark Box) and novel object investigation of juveniles indicating an inhibitory role for the juvenile ovary; social approach was unaffected. In contrast, adult ovariectomy and castration (both prepubertal and adult) had no effect on any behavioral measure. Experiment 3 tested whether rearing hamsters in a short day length (SD), which delays puberty in this species, extends the interval of juvenile ovarian inhibition on exploration and novelty seeking. We also tested whether provision of estradiol reverses the effects of prepubertal ovariectomy. Hormonal manipulations and behavioral tests of Experiment 3 were conducted at ages when long day-reared hamsters are adult (as in Experiment 2), but SD-reared hamsters remain reproductively immature. Ovariectomy again increased exploration in the SD-reared juveniles despite the older age of surgery and testing. Estradiol treatment had no effect. These findings reveal a novel role for the juvenile ovary in exploration and novelty seeking that is unlikely to be mediated exclusively by estradiol.
Collapse
Affiliation(s)
- R F Kyne
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY, USA; Neuroscience Program, University at Buffalo SUNY, Buffalo, NY, USA.
| | - A R Barrett
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY, USA.
| | - L M Brown
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY, USA.
| | - M J Paul
- Department of Psychology, University at Buffalo SUNY, Buffalo, NY, USA; Neuroscience Program, University at Buffalo SUNY, Buffalo, NY, USA; Evolution, Ecology, and Behavior Program, University at Buffalo SUNY, Buffalo, NY, USA.
| |
Collapse
|
23
|
Cisternas CD, Cortes LR, Bruggeman EC, Yao B, Forger NG. Developmental changes and sex differences in DNA methylation and demethylation in hypothalamic regions of the mouse brain. Epigenetics 2019; 15:72-84. [PMID: 31378140 DOI: 10.1080/15592294.2019.1649528] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
DNA methylation is dynamically modulated during postnatal brain development, and plays a key role in neuronal lineage commitment. This epigenetic mark has also recently been implicated in the development of neural sex differences, many of which are found in the hypothalamus. The level of DNA methylation depends on a balance between the placement of methyl marks by DNA methyltransferases (Dnmts) and their removal, which is catalyzed by ten-eleven translocation (Tet) methylcytosine dioxygenases. Here, we examined developmental changes and sex differences in the expression of Tet and Dnmt enzymes from birth to adulthood in two hypothalamic regions (the preoptic area and ventromedial nucleus) and the hippocampus of mice. We found highest expression of all Tet enzymes (Tet1, Tet2, Tet3) and Dnmts (Dnmt1, Dnmt3a, Dnmt3b) in newborns, despite the fact that global methylation and hydroxymethylation were at their lowest levels at birth. Expression of the Dnmt co-activator, Dnmt3l, followed a pattern opposite to that of the canonical Dnmts (i.e., was very low in newborns and increased with age). Tet enzyme activity was much higher at birth than at weaning in both the hypothalamus and hippocampus, mirroring developmental changes in gene expression. Sex differences in Tet enzyme expression were seen in all brain regions examined during the first week of life, whereas Dnmt expression was more balanced between the sexes. Neonatal testosterone treatment of females only partially masculinized enzyme expression. Thus, Tet expression and activity are elevated during neonatal brain development, and may play important roles in sexual differentiation of the brain.
Collapse
Affiliation(s)
- Carla D Cisternas
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| | - Laura R Cortes
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| | - Emily C Bruggeman
- Department of Human Genetics, Emory School of Medicine, Atlanta, GA, USA
| | - Bing Yao
- Department of Human Genetics, Emory School of Medicine, Atlanta, GA, USA
| | - Nancy G Forger
- Neuroscience Institute and Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
24
|
Topper VY, Reilly MP, Wagner LM, Thompson LM, Gillette R, Crews D, Gore AC. Social and neuromolecular phenotypes are programmed by prenatal exposures to endocrine-disrupting chemicals. Mol Cell Endocrinol 2019; 479:133-146. [PMID: 30287398 PMCID: PMC6263824 DOI: 10.1016/j.mce.2018.09.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/25/2018] [Accepted: 09/28/2018] [Indexed: 01/09/2023]
Abstract
Exposures to endocrine-disrupting chemicals (EDCs) affect the development of hormone-sensitive neural circuits, the proper organization of which are necessary for the manifestation of appropriate adult social and sexual behaviors. We examined whether prenatal exposure to polychlorinated biphenyls (PCBs), a family of ubiquitous industrial contaminants detectable in virtually all humans and wildlife, caused changes in sexually-dimorphic social interactions and communications, and profiled the underlying neuromolecular phenotype. Rats were treated with a PCB commercial mixture, Aroclor 1221 (A1221), estradiol benzoate (EB) as a positive control for estrogenic effects of A1221, or the vehicle (4% DMSO), on embryonic day (E) 16 and 18. In adult F1 offspring, we first conducted tests of ultrasonic vocalization (USV) calls in a sociosexual context as a measure of motivated communications. Numbers of certain USV call types were significantly increased by prenatal treatment with A1221 in males, and decreased by EB in females. In a test of sociosexual preference for a hormone-vs. a non-hormone-primed opposite sex conspecific, male (but not female) nose-touching with opposite-sex rats was significantly diminished by EDCs. Gene expression profiling was conducted in two brain regions that are part of the social decision-making network in the brain: the medial preoptic nucleus (MPN) and the ventromedial nucleus (VMN). In both regions, many more genes were affected by A1221 or EB in females than males. In female MPN, A1221 changed expression of steroid hormone receptor and neuropeptide genes (e.g., Ar, Esr1, Esr2, and Kiss1). In male MPN, only Per2 was affected by A1221. The VMN had a number of genes affected by EB compared to vehicle (females: Kiss1, Kiss1r, Pgr; males: Crh) but not A1221. These differences between EB and A1221 indicate that the mechanism of action of A1221 goes beyond estrogenic pathways. These data show sex-specific effects of prenatal PCBs on adult behaviors and the neuromolecular phenotype.
Collapse
Affiliation(s)
- Viktoria Y Topper
- The Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Michael P Reilly
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lauren M Wagner
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lindsay M Thompson
- Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ross Gillette
- The Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - David Crews
- The Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA; Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea C Gore
- The Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA; Division of Pharmacology and Toxicology, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
25
|
Neurobiological characteristics underlying metabolic differences between males and females. Prog Neurobiol 2018; 176:18-32. [PMID: 30194984 DOI: 10.1016/j.pneurobio.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/22/2018] [Accepted: 09/01/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.
Collapse
|
26
|
Bell MR. Comparing Postnatal Development of Gonadal Hormones and Associated Social Behaviors in Rats, Mice, and Humans. Endocrinology 2018; 159:2596-2613. [PMID: 29767714 PMCID: PMC6692888 DOI: 10.1210/en.2018-00220] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022]
Abstract
Postnatal development includes dramatic changes in gonadal hormones and the many social behaviors they help regulate, both in rodents and humans. Parental care-seeking is the most salient social interaction in neonates and infants, play and prosocial behaviors are commonly studied in juveniles, and the development of aggression and sexual behavior begins in peripubertal stages but continues through late adolescence into adulthood. Although parental behaviors are shown after reproductive success in adulthood, alloparenting behaviors are actually high in juveniles as well. These behaviors are sensitive to both early-life organizational effects of gonadal hormones and later-life activational regulation. However, changes in circulating gonadal hormones and the display of the previous behaviors over development differ between rats, mice, and humans. These endpoints are of interest to endocrinologist, toxicologists, and neuroscientists because of their relevance to mental health disorders and their vulnerability to effects of endocrine-disrupting chemical exposure. As such, the goal of this mini-review is to succinctly describe and relate the postnatal development of gonadal hormones and social behaviors to each other, over time, and across animal models. Ideally, this will help identify appropriate animal models and age ranges for continued study of both normative development and in contexts of environmental disruption.
Collapse
Affiliation(s)
- Margaret R Bell
- Department of Biological Sciences, DePaul University, Chicago, Illinois
- Department of Health Sciences, DePaul University, Chicago, Illinois
| |
Collapse
|
27
|
Gore AC, Holley AM, Crews D. Mate choice, sexual selection, and endocrine-disrupting chemicals. Horm Behav 2018; 101:3-12. [PMID: 28888817 PMCID: PMC5845777 DOI: 10.1016/j.yhbeh.2017.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/26/2017] [Accepted: 09/05/2017] [Indexed: 12/30/2022]
Abstract
Humans have disproportionately affected the habitat and survival of species through environmental contamination. Important among these anthropogenic influences is the proliferation of organic chemicals, some of which perturb hormone systems, the latter referred to as endocrine-disrupting chemicals (EDCs). EDCs are widespread in the environment and affect all levels of reproduction, including development of reproductive organs, hormone release and regulation through the life cycle, the development of secondary sexual characteristics, and the maturation and maintenance of adult physiology and behavior. However, what is not well-known is how the confluence of EDC actions on the manifestation of morphological and behavioral sexual traits influences mate choice, a process that requires the reciprocal evaluation of and/or acceptance of a sexual partner. Moreover, the outcomes of EDC-induced perturbations are likely to influence sexual selection; yet this has rarely been directly tested. Here, we provide background on the development and manifestation of sexual traits, reproductive competence, and the neurobiology of sexual behavior, and evidence for their perturbation by EDCs. Selection acts on individuals, with the consequences manifest in populations, and we discuss the implications for EDC contamination of these processes, and the future of species.
Collapse
Affiliation(s)
- Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, USA.
| | - Amanda M Holley
- Division of Pharmacology and Toxicology, College of Pharmacy, USA; Department of Integrative Biology, College of Natural Sciences, USA
| | - David Crews
- Department of Integrative Biology, College of Natural Sciences, USA.
| |
Collapse
|
28
|
Manoli DS, Tollkuhn J. Gene regulatory mechanisms underlying sex differences in brain development and psychiatric disease. Ann N Y Acad Sci 2018; 1420:26-45. [PMID: 29363776 PMCID: PMC5991992 DOI: 10.1111/nyas.13564] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/26/2017] [Accepted: 11/01/2017] [Indexed: 12/12/2022]
Abstract
The sexual differentiation of the mammalian nervous system requires the precise coordination of the temporal and spatial regulation of gene expression in diverse cell types. Sex hormones act at multiple developmental time points to specify sex-typical differentiation during embryonic and early development and to coordinate subsequent responses to gonadal hormones later in life by establishing sex-typical patterns of epigenetic modifications across the genome. Thus, mutations associated with neuropsychiatric conditions may result in sexually dimorphic symptoms by acting on different neural substrates or chromatin landscapes in males and females. Finally, as stress hormone signaling may directly alter the molecular machinery that interacts with sex hormone receptors to regulate gene expression, the contribution of chronic stress to the pathogenesis or presentation of mental illness may be additionally different between the sexes. Here, we review the mechanisms that contribute to sexual differentiation in the mammalian nervous system and consider some of the implications of these processes for sex differences in neuropsychiatric conditions.
Collapse
Affiliation(s)
- Devanand S. Manoli
- Department of Psychiatry and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, California
| | | |
Collapse
|
29
|
Reilly MP, Weeks CD, Crews D, Gore AC. Application of a novel social choice paradigm to assess effects of prenatal endocrine-disrupting chemical exposure in rats (Rattus norvegicus). ACTA ACUST UNITED AC 2018; 132:253-267. [PMID: 29683687 DOI: 10.1037/com0000103] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Endocrine-disrupting chemical (EDC) exposures during critical periods of gestation cause long-lasting behavioral effects, presumably by disturbing hormonal organization of the brain. Among such EDCs are polychlorinated biphenyls (PCBs), a class of industrial chemicals. PCB exposure in utero leads to alterations in mating behaviors and other sexually dimorphic social interactions in rats. Many of the previous studies on social behavior gave the experimental animal a single or binary choice. This study applied a more complex behavioral apparatus, an X-shaped Plexiglas apparatus (FourPlex), that enabled an experimental animal exposed to PCBs or a vehicle to distinguish and choose among 4 stimulus animals of the same or opposite sex, and of different hormonal status. We found that rats were able to differentiate among the stimuli in the FourPlex and showed the expected preference for an opposite sex, hormone-treated rat, particularly for behaviors conducted in proximity. Prenatal treatment caused subtle shifts in behavior toward stimulus rats in the FourPlex; more robust effects were seen for the sexual dimorphisms in behavior. Importantly, the results differ from our previous results of a simple binary choice model, showing that how an animal behaves in a more complex social paradigm does not predict the outcome in a simple choice model, and vice versa. (PsycINFO Database Record
Collapse
Affiliation(s)
- Michael P Reilly
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin
| | - Connor D Weeks
- College of Natural Sciences, The University of Texas at Austin
| | - David Crews
- Section of Integrative Biology, The University of Texas at Austin
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin
| |
Collapse
|
30
|
Hou H, Uusküla-Reimand L, Makarem M, Corre C, Saleh S, Metcalf A, Goldenberg A, Palmert MR, Wilson MD. Gene expression profiling of puberty-associated genes reveals abundant tissue and sex-specific changes across postnatal development. Hum Mol Genet 2018; 26:3585-3599. [PMID: 28911201 DOI: 10.1093/hmg/ddx246] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
The timing of human puberty is highly variable, sexually dimorphic, and associated with adverse health outcomes. Over 20 genes carrying rare mutations have been identified in known pubertal disorders, many of which encode critical components of the hypothalamic-pituitary-gonadal (HPG) axis. Recent genome-wide association studies (GWAS) have identified more than 100 candidate genes at loci associated with age at menarche or voice breaking in males. We know little about the spatial, temporal or postnatal expression patterns of the majority of these puberty-associated genes. Using a high-throughput and sensitive microfluidic quantitative PCR strategy, we profiled the gene expression patterns of the mouse orthologs of 178 puberty-associated genes in male and female mouse HPG axis tissues, the pineal gland, and the liver at five postnatal ages spanning the pubertal transition. The most dynamic gene expression changes were observed prior to puberty in all tissues. We detected known and novel tissue-enhanced gene expression patterns, with the hypothalamus expressing the largest number of the puberty-associated genes. Notably, over 40 puberty-associated genes in the pituitary gland showed sex-biased gene expression, most of which occurred peri-puberty. These sex-biased genes included the orthologs of candidate genes at GWAS loci that show sex-discordant effects on pubertal timing. Our findings provide new insight into the expression of puberty-associated genes and support the possibility that the pituitary plays a role in determining sex differences in the timing of puberty.
Collapse
Affiliation(s)
- Huayun Hou
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Liis Uusküla-Reimand
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Gene Technology, Tallinn University of Technology, 12616 Tallinn, Estonia
| | - Maisam Makarem
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Christina Corre
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Shems Saleh
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Computer Science, University of Toronto, Toronto, ON M5S 2E5, Canada
| | - Ariane Metcalf
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Anna Goldenberg
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Computer Science, University of Toronto, Toronto, ON M5S 2E5, Canada
| | - Mark R Palmert
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Division of Endocrinology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,Departments of Paediatrics and Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael D Wilson
- Program in Genetics and Genome Biology, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
31
|
Picut CA, Ziejewski MK, Stanislaus D. Comparative Aspects of Pre- and Postnatal Development of the Male Reproductive System. Birth Defects Res 2017; 110:190-227. [PMID: 29063715 DOI: 10.1002/bdr2.1133] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/29/2017] [Accepted: 08/30/2017] [Indexed: 01/01/2023]
Abstract
This review describes pre- and postnatal development of the male reproductive system in humans and laboratory animals, and highlights species differences in the timing and control of hormonal and morphologic events. Major differences are that the fetal testis is dependent on gonadotropins in humans, but is independent of such in rats; humans have an extended postnatal quiescent period, whereas rats exhibit no quiescence; and events such as secretion by the prostate and seminal vesicles, testicular descent, and the appearance of spermatogonia are all prenatal events in humans, but are postnatal events in rats. Major differences in the timing of the developmental sequence between rats and humans include: gonocyte transformation period (rat: postnatal day 0-9; human: includes gestational week 22 to 9 months of age); masculinization programming window (rat: gestational day 15.5-17.5; human: gestational week 9-14); and mini-puberty (rat: 0-6 hr after birth; human: 3-6 months of age). Endocrine disruptors can cause unique lesions in the prenatal and early postnatal testis; therefore, it is important to consider the differences in the timing of the developmental sequence when designing preclinical studies as identification of windows of sensitivity for endocrine disruption or toxicants will aid in interpretation of results and provide clues to a mode of action. Birth Defects Research 110:190-227, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Catherine A Picut
- Charles River Laboratories, Pathology Associates, Durham, North Carolina
| | - Mary K Ziejewski
- GlaxoSmithKline Research & Development, King of Prussia, Pennsylvania
| | - D Stanislaus
- GlaxoSmithKline Research & Development, King of Prussia, Pennsylvania
| |
Collapse
|
32
|
Roepke TA, Yasrebi A, Villalobos A, Krumm EA, Yang JA, Mamounis KJ. Loss of ERα partially reverses the effects of maternal high-fat diet on energy homeostasis in female mice. Sci Rep 2017; 7:6381. [PMID: 28743985 PMCID: PMC5526977 DOI: 10.1038/s41598-017-06560-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/13/2017] [Indexed: 11/18/2022] Open
Abstract
Maternal high-fat diet (HFD) alters hypothalamic developmental programming and disrupts offspring energy homeostasis in rodents. 17β-estradiol (E2) also influences hypothalamic programming through estrogen receptor (ER) α. Therefore, we hypothesized that females lacking ERα would be more susceptible to maternal HFD. To address this question, heterozygous ERα knockout (WT/KO) dams were fed a control breeder chow diet (25% fat) or a semi-purified HFD (45% fat) 4 weeks prior to mating with WT/KO males or heterozygous males with an ERα DNA-binding domain mutation knocked in (WT/KI) to produce WT, ERα KO, or ERα KIKO females lacking ERE-dependent ERα signaling. Maternal HFD increased body weight in WT and KIKO, in part, due to increased adiposity and daytime carbohydrate utilization in WT and KIKO, while increasing nighttime fat utilization in KO. Maternal HFD also increased plasma leptin, IL-6, and MCP-1 in WT and increased arcuate expression of Kiss1 and Esr1 (ERα) and liver expression of G6pc and Pepck in WT and KIKO. Contrary to our hypothesis, these data suggest that loss of ERα signaling blocks the influence of maternal HFD on energy homeostasis, inflammation, and hypothalamic and liver gene expression and that restoration of ERE-independent ERα signaling partially reestablishes susceptibility to maternal HFD.
Collapse
Affiliation(s)
- Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA. .,New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Alejandra Villalobos
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Elizabeth A Krumm
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Reproductive Medicine, University of California, San Diego, San Diego, CA 92103, USA
| | - Kyle J Mamounis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
33
|
Meerts SH, Anderson KS, Farry-Thorn ME, Johnson EG, Taxier L. Prepubertal ovariectomy modulates paced mating behavior but not sexual preference or conditioned place preference for mating in female rats. Physiol Behav 2017; 171:142-148. [PMID: 28082246 DOI: 10.1016/j.physbeh.2017.01.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/07/2017] [Accepted: 01/07/2017] [Indexed: 11/24/2022]
Abstract
The present study investigated whether the presence or absence of peripubertal ovarian hormones affects sexual preference and conditioned place preference for paced mating in adult female rats primed with 10μg estradiol benzoate and 1mg progesterone. Ovariectomy (OVX) occurred either before or after pubertal development, and 4weeks later rats began a series of behavioral tests. Rats with ovaries removed before the pubertal timeframe (Prepubertal OVX) were more active, more likely to withdrawal from the male compartment, and did not discriminate between mounts and intromissions during paced mating relative to rats with ovaries during puberty (Adult OVX). Both Adult OVX and Prepubertal OVX rats showed a higher preference for the male when hormone primed vs. oil treated and a conditioned place preference for paced mating behavior. The results of the present study demonstrate that some, but not all, aspects of female sexual behavior require ovarian hormones during puberty.
Collapse
Affiliation(s)
- Sarah H Meerts
- Department of Psychology, Carleton College, Northfield, MN 55057, United States.
| | - Kelly S Anderson
- Department of Psychology, Carleton College, Northfield, MN 55057, United States
| | - Molly E Farry-Thorn
- Department of Psychology, Carleton College, Northfield, MN 55057, United States
| | - Elliott G Johnson
- Department of Psychology, Carleton College, Northfield, MN 55057, United States
| | - Lisa Taxier
- Department of Psychology, Carleton College, Northfield, MN 55057, United States
| |
Collapse
|
34
|
Hodes GE, Walker DM, Labonté B, Nestler EJ, Russo SJ. Understanding the epigenetic basis of sex differences in depression. J Neurosci Res 2017; 95:692-702. [PMID: 27870456 PMCID: PMC5130105 DOI: 10.1002/jnr.23876] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/13/2016] [Accepted: 07/14/2016] [Indexed: 12/27/2022]
Abstract
Epigenetics refers to potentially heritable processes that can mediate both lasting and transient changes in gene expression in the absence of genome sequence alterations. The field of epigenetics has introduced a novel understanding of the mechanisms through which the environment can shape an individual and potentially its offspring. This Mini-Review examines the current literature exploring the role of epigenetics in the development of mood disorders such as depression. Depression is twofold more common in females, yet the majority of preclinical research has been conducted exclusively in male subjects. Here we discuss what is known about sex differences in epigenetic regulation and function and how this may contribute to the etiology and onset of mood disorders. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Georgia E. Hodes
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Deena M. Walker
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Benoit Labonté
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Eric J. Nestler
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Scott J. Russo
- Fishberg Department of Neuroscience and Freidman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
35
|
Walker DM, Gore AC. Epigenetic impacts of endocrine disruptors in the brain. Front Neuroendocrinol 2017; 44:1-26. [PMID: 27663243 PMCID: PMC5429819 DOI: 10.1016/j.yfrne.2016.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/05/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
The acquisition of reproductive competence is organized and activated by steroid hormones acting upon the hypothalamus during critical windows of development. This review describes the potential role of epigenetic processes, particularly DNA methylation, in the regulation of sexual differentiation of the hypothalamus by hormones. We examine disruption of these processes by endocrine-disrupting chemicals (EDCs) in an age-, sex-, and region-specific manner, focusing on how perinatal EDCs act through epigenetic mechanisms to reprogram DNA methylation and sex steroid hormone receptor expression throughout life. These receptors are necessary for brain sexual differentiation and their altered expression may underlie disrupted reproductive physiology and behavior. Finally, we review the literature on histone modifications and non-coding RNA involvement in brain sexual differentiation and their perturbation by EDCs. By putting these data into a sex and developmental context we conclude that perinatal EDC exposure alters the developmental trajectory of reproductive neuroendocrine systems in a sex-specific manner.
Collapse
Affiliation(s)
- Deena M Walker
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1065, New York, NY 10029, USA.
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Institute for Cellular and Molecular Biology, and The University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
36
|
Onaolapo OJ, Onaolapo AY, Omololu TA, Oludimu AT, Segun-Busari T, Omoleke T. Exogenous Testosterone, Aging, and Changes in Behavioral Response of Gonadally Intact Male Mice. J Exp Neurosci 2016; 10:59-70. [PMID: 27158222 PMCID: PMC4854217 DOI: 10.4137/jen.s39042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/06/2016] [Accepted: 03/18/2016] [Indexed: 01/23/2023] Open
Abstract
This study tested the hypothesis that aging significantly affects the influence of exogenous testosterone on neurobehavior in gonadally intact male mice. Groups of prepubertal and aged male mice received daily vehicle or testosterone propionate (TP; 2.5 or 5.0 mg/kg intraperitoneal [i.p.]) for 21 days. Behaviors were assessed on days 1 and 21. Weight gain was significant in prepubertal mice. Locomotion and rearing increased in prepubertal mice after first dose and decreased after last dose of TP. Rearing was suppressed in aged mice throughout. Suppression of grooming occurred in both age groups at day 21. Significant increase in working memory in both age groups was seen in the radial-arm maze (at specific doses) and in prepubertal mice in the Y-maze. Elevated plus maze test showed mixed anxiolytic/anxiogenic effects. Aged mice had higher serum testosterone. In conclusion, age is an important determinant for the influence of exogenous testosterone on behavior in gonadally intact male mice.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Oshogbo, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Tope A Omololu
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adedunke T Oludimu
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Toluwalase Segun-Busari
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Taofeeq Omoleke
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
37
|
Clarkson J, Herbison AE. Hypothalamic control of the male neonatal testosterone surge. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150115. [PMID: 26833836 PMCID: PMC4785901 DOI: 10.1098/rstb.2015.0115] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2015] [Indexed: 11/12/2022] Open
Abstract
Sex differences in brain neuroanatomy and neurophysiology underpin considerable physiological and behavioural differences between females and males. Sexual differentiation of the brain is regulated by testosterone secreted by the testes predominantly during embryogenesis in humans and the neonatal period in rodents. Despite huge advances in understanding how testosterone, and its metabolite oestradiol, sexually differentiate the brain, little is known about the mechanism that actually generates the male-specific neonatal testosterone surge. This review examines the evidence for the role of the hypothalamus, and particularly the gonadotropin-releasing hormone (GnRH) neurons, in generating the neonatal testosterone surge in rodents and primates. Kisspeptin-GPR54 signalling is well established as a potent and critical regulator of GnRH neuron activity during puberty and adulthood, and we argue here for an equally important role at birth in driving the male-specific neonatal testosterone surge in rodents. The presence of a male-specific population of preoptic area kisspeptin neurons that appear transiently in the perinatal period provide one possible source of kisspeptin drive to neonatal GnRH neurons in the mouse.
Collapse
Affiliation(s)
- Jenny Clarkson
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Allan E Herbison
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
38
|
Purves-Tyson TD, Allen K, Fung S, Rothmond D, Noble PL, Handelsman DJ, Shannon Weickert C. Adolescent testosterone influences BDNF and TrkB mRNA and neurotrophin-interneuron marker relationships in mammalian frontal cortex. Schizophr Res 2015; 168:661-70. [PMID: 26088421 DOI: 10.1016/j.schres.2015.05.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/11/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
Late adolescence in males is a period of increased susceptibility for the onset of schizophrenia, coinciding with increased circulating testosterone. The cognitive deficits prevalent in schizophrenia may be related to unhealthy cortical interneurons, which are trophically dependent on brain derived neurotrophic factor. We investigated, under conditions of depleted (monkey and rat) and replaced (rat) testosterone over adolescence, changes in gene expression of cortical BDNF and TrkB transcripts and interneuron markers and the relationships between these mRNAs and circulating testosterone. Testosterone removal by gonadectomy reduced gene expression of some BDNF transcripts in monkey and rat frontal cortices and the BDNF mRNA reduction was prevented by testosterone replacement. In rat, testosterone replacement increased the potential for classical TrkB signalling by increasing the full length to truncated TrkB mRNA ratio, whereas in the monkey cortex, circulating testosterone was negatively correlated with the TrkB full length/truncated mRNA ratio. We did not identify changes in interneuron gene expression in monkey frontal cortex in response to gonadectomy, and in rat, we showed that only somatostatin mRNA was decreased by gonadectomy but not restored by testosterone replacement. We identified complex and possibly species-specific, relationships between BDNF/TrkB gene expression and interneuron marker gene expression that appear to be dependent on the presence of testosterone at adolescence in rat and monkey frontal cortices. Taken together, our findings suggest there are dynamic relationships between BDNF/TrkB and interneuron markers that are dependent on the presence of testosterone but that this may not be a straightforward increase in testosterone leading to changes in BDNF/TrkB that contributes to interneuron health.
Collapse
Affiliation(s)
- Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Medical Sciences, University of New South Wales, Sydney 2031, Australia
| | - Katherine Allen
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Samantha Fung
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| | - Debora Rothmond
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia
| | | | | | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney 2021, Australia; Neuroscience Research Australia, Sydney 2031, Australia; School of Psychiatry, University of New South Wales, Sydney 2031, Australia
| |
Collapse
|
39
|
Topper VY, Walker DM, Gore AC. Sexually dimorphic effects of gestational endocrine-disrupting chemicals on microRNA expression in the developing rat hypothalamus. Mol Cell Endocrinol 2015; 414:42-52. [PMID: 26190835 PMCID: PMC4553128 DOI: 10.1016/j.mce.2015.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/30/2015] [Accepted: 07/14/2015] [Indexed: 12/11/2022]
Abstract
This study examined developmental changes and sexual dimorphisms in hypothalamic microRNAs, and whether gestational exposures to environmental endocrine-disrupting chemicals (EDCs) altered their expression patterns. Pregnant rat dams were treated on gestational days 16 and 18 with vehicle, estradiol benzoate, or a mixture of polychlorinated biphenyls. Male and female offspring were euthanized on postnatal days (P) 15, 30, 45, or 90, and microRNA and mRNA targets were quantified in the medial preoptic nucleus (MPN) and ventromedial nucleus (VMN) of the hypothalamus. MicroRNAs showed robust developmental changes in both regions, and were sexually dimorphic in the MPN, but not VMN. Importantly, microRNAs in females were up-regulated by EDCs at P30, and down-regulated in males at P90. Few changes in mRNAs were found. Thus, hypothalamic microRNAs are sensitive to prenatal EDC treatment in a sex-, developmental age-, and brain region-specific manner.
Collapse
Affiliation(s)
- Viktoria Y Topper
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Deena M Walker
- Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea C Gore
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA; Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
40
|
Naulé L, Robert V, Parmentier C, Martini M, Keller M, Cohen-Solal M, Hardin-Pouzet H, Grange-Messent V, Franceschini I, Mhaouty-Kodja S. Delayed pubertal onset and prepubertal Kiss1 expression in female mice lacking central oestrogen receptor beta. Hum Mol Genet 2015; 24:7326-38. [PMID: 26464488 DOI: 10.1093/hmg/ddv430] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/07/2015] [Indexed: 01/02/2023] Open
Abstract
Ovarian oestradiol is essential for pubertal maturation and adult physiology of the female reproductive axis. It acts at central and peripheral sites through two main oestrogen receptors (ER) α and β. Here we investigate the role of ERβ on central effects of oestradiol, by generating a mouse line specifically lacking the ERβ gene in neuronal and glial cells. Central ERβ deletion delays the age at vaginal opening and first oestrous and reduces uterine weight without affecting body growth. Analysis of factors necessary for pubertal progression shows reduced levels of Kiss1 transcripts at postnatal (P) day 25 in the preoptic area, but not in the mediobasal hypothalamus (MBH) of mutant females. In agreement with these data, the number of kisspeptin-immunoreactive neurons was decreased by 57-72% in the three subdivisions of the rostral periventricular area of the third ventricle (RP3V), whereas the density of kisspeptin-immunoreactive fibres was unchanged in the arcuate nucleus of mutant mice. These alterations do not involve changes in ERα mRNAs in the preoptic area and protein levels in the RP3V. The number and distribution of GnRH-immunoreactive cells were unaffected, but gonadotropin-releasing hormone (GnRH) transcript levels were higher in the P25 preoptic area of mutants. At adulthood, mutant females have normal oestrous cyclicity, kisspeptin system and exhibit unaltered sexual behaviour. They display, however, reduced ovary weight and increased anxiety-related behaviour during the follicular phase. This argues for the specific involvement of central ERβ in the regulation of pubertal onset in female reproduction, possibly through prepubertal induction of kisspeptin expression in the RP3V.
Collapse
Affiliation(s)
- Lydie Naulé
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Vincent Robert
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Caroline Parmentier
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Mariangela Martini
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Matthieu Keller
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Martine Cohen-Solal
- Inserm U1132 and university Paris-Diderot, Hospital Lariboisière, Paris, France
| | - Hélène Hardin-Pouzet
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Valérie Grange-Messent
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France
| | - Isabelle Franceschini
- Institut National de la Recherche Agronomique, UMR 85, Nouzilly, France, Centre National de la Recherche Scientifique, UMR 7247, Nouzilly, France, Université François Rabelais, Tours, France, Institut Français du Cheval et de l'Equitation, Nouzilly, France and
| | - Sakina Mhaouty-Kodja
- Neuroscience Paris Seine, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche (UMR) S1130, Centre National de la Recherche Scientifique, UMR 8246, Université P. et M. Curie, Paris, France, Sorbonne Universités, Université P. et M. Curie UM CR18, Université Paris 06, Paris, France,
| |
Collapse
|
41
|
Yin W, Maguire SM, Pham B, Garcia AN, Dang NV, Liang J, Wolfe A, Hofmann HA, Gore AC. Testing the Critical Window Hypothesis of Timing and Duration of Estradiol Treatment on Hypothalamic Gene Networks in Reproductively Mature and Aging Female Rats. Endocrinology 2015; 156:2918-33. [PMID: 26018250 PMCID: PMC4511137 DOI: 10.1210/en.2015-1032] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022]
Abstract
At menopause, the dramatic loss of ovarian estradiol (E2) necessitates the adaptation of estrogen-sensitive neurons in the hypothalamus to an estrogen-depleted environment. We developed a rat model to test the "critical window" hypothesis of the effects of timing and duration of E2 treatment after deprivation on the hypothalamic neuronal gene network in the arcuate nucleus and the medial preoptic area. Rats at 2 ages (reproductively mature or aging) were ovariectomized and given E2 or vehicle replacement regimes of differing timing and duration. Using a 48-gene quantitative low-density PCR array and weighted gene coexpression network analysis, we identified gene modules differentially regulated by age, timing, and duration of E2 treatment. Of particular interest, E2 status differentially affected suites of genes in the hypothalamus involved in energy balance, circadian rhythms, and reproduction. In fact, E2 status was the dominant factor in determining gene modules and hormone levels; age, timing, and duration had more subtle effects. Our results highlight the plasticity of hypothalamic neuroendocrine systems during reproductive aging and its surprising ability to adapt to diverse E2 replacement regimes.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Sean M Maguire
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Brian Pham
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Alexandra N Garcia
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Nguyen-Vy Dang
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Jingya Liang
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Andrew Wolfe
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Hans A Hofmann
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| | - Andrea C Gore
- Division of Pharmacology and Toxicology (W.Y., B.P., N.-V.D., J.L., A.C.G.), Departments of Integrative Biology (S.M.M., H.A.H.) and Psychology (A.N.G., A.C.G.), and Institute for Neuroscience (H.A.H., A.C.G.), The University of Texas at Austin, Austin, Texas 78712; and Johns Hopkins University School of Medicine (A.W.), Baltimore, Maryland 21287
| |
Collapse
|
42
|
Reilly MP, Weeks CD, Topper VY, Thompson LM, Crews D, Gore AC. The effects of prenatal PCBs on adult social behavior in rats. Horm Behav 2015; 73:47-55. [PMID: 26093262 PMCID: PMC4546928 DOI: 10.1016/j.yhbeh.2015.06.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 05/27/2015] [Accepted: 06/05/2015] [Indexed: 11/15/2022]
Abstract
Endocrine disrupting chemical (EDC) exposures during critical periods of development may influence neuronal development and the manifestation of sexually dimorphic sociability and social novelty behaviors in adulthood. In this study, we assessed the effects of gestational exposure to PCBs on the social behavior of males and females later in adulthood. A weakly estrogenic PCB mixture, Aroclor 1221 (A1221, 0.5 or 1mg/kg) was administered to pregnant Sprague-Dawley rat dams. Both a positive control (estradiol benzoate; EB, 50μg/kg) and negative control (dimethylsulfoxide; DMSO in sesame oil vehicle) were similarly administered to separate sets of dams. The sexes responded differently in two tasks essential to sociality. Using a three-chamber apparatus that contained a caged, same-sex, gonadectomized stimulus animal and an empty stimulus cage, we found that both sexes showed a strong preference for affiliating with a stimulus animal (vs. an empty cage), an effect that was much more pronounced in the males. In the second task, a novel and a familiar stimulus animal were caged at opposite ends of the same apparatus. Females displayed a higher degree of novelty preference than the males. During both tests, females had significantly higher social approach behaviors while male engaged in significantly more interactive behaviors with the conspecific. Of particular interest, males born of dams that received prenatal A1221 (0.5mg/kg) exhibited an overall decrease in nose-to-nose investigations. These behavioral data suggest that the males are more sensitive to A1221 treatment than are females. In addition to behavioral analysis, serum corticosterone was measured. Females born of dams treated with A1221 (0.5mg/kg) had significantly higher concentrations of corticosterone than the DMSO female group; males were unaffected. Females also had significantly higher corticosterone concentrations than did males. Overall, our results suggest that the effects of gestational exposure to PCBs on adult social behavior are relatively limited within this particular paradigm.
Collapse
Affiliation(s)
- Michael P Reilly
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Connor D Weeks
- College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Viktoria Y Topper
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lindsay M Thompson
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - David Crews
- Section of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea C Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
43
|
Purves-Tyson TD, Boerrigter D, Allen K, Zavitsanou K, Karl T, Djunaidi V, Double KL, Desai R, Handelsman DJ, Weickert CS. Testosterone attenuates and the selective estrogen receptor modulator, raloxifene, potentiates amphetamine-induced locomotion in male rats. Horm Behav 2015; 70:73-84. [PMID: 25747465 DOI: 10.1016/j.yhbeh.2015.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 01/16/2015] [Accepted: 02/28/2015] [Indexed: 11/17/2022]
Abstract
Although sex steroids are known to modulate brain dopamine, it is still unclear how testosterone modifies locomotor behaviour controlled, at least in part, by striatal dopamine in adolescent males. Our previous work suggests that increasing testosterone during adolescence may bias midbrain neurons to synthesise more dopamine. We hypothesised that baseline and amphetamine-induced locomotion would differ in adult males depending on testosterone exposure during adolescence. We hypothesised that concomitant stimulation of estrogen receptor signaling, through a selective estrogen receptor modulator (SERM), raloxifene, can counter testosterone effects on locomotion. Male Sprague-Dawley rats at postnatal day 45 were gonadectomised (G) or sham-operated (S) prior to the typical adolescent testosterone increase. Gonadectomised rats were either given testosterone replacement (T) or blank implants (B) for six weeks and sham-operated (i.e. intact or endogenous testosterone group) were given blank implants. Subgroups of sham-operated, gonadectomised and gonadectomised/testosterone-replaced rats were treated with raloxifene (R, 5mg/kg) or vehicle (V), daily for the final four weeks. There were six groups (SBV, GBV, GTV, SBR, GBR, GTR). Saline and amphetamine-induced (1.25mg/kg) locomotion in the open field was measured at PND85. Gonadectomy increased amphetamine-induced locomotion compared to rats with endogenous or with exogenous testosterone. Raloxifene increased amphetamine-induced locomotion in rats with either endogenous or exogenous testosterone. Amphetamine-induced locomotion was negatively correlated with testosterone and this relationship was abolished by raloxifene. Lack of testosterone during adolescence potentiates and testosterone exposure during adolescence attenuates amphetamine-induced locomotion. Treatment with raloxifene appears to potentiate amphetamine-induced locomotion and to have an opposite effect to that of testosterone in male rats.
Collapse
Affiliation(s)
- Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia.
| | - Danny Boerrigter
- Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia
| | - Katherine Allen
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Katerina Zavitsanou
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Tim Karl
- Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia
| | - Vanezha Djunaidi
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Medical Sciences, Faculty of Medicine, University of New South Wales, Australia
| | - Kay L Double
- Discipline of Biomedical Science, School of Medical Sciences, Sydney Medical School, University of Sydney, Australia
| | - Reena Desai
- ANZAC Research Institute, University of Sydney, Concord Hospital, Australia
| | - David J Handelsman
- ANZAC Research Institute, University of Sydney, Concord Hospital, Australia
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Sydney, Australia; School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
44
|
Allen KM, Purves-Tyson TD, Fung SJ, Shannon Weickert C. The effect of adolescent testosterone on hippocampal BDNF and TrkB mRNA expression: relationship with cell proliferation. BMC Neurosci 2015; 16:4. [PMID: 25886766 PMCID: PMC4367905 DOI: 10.1186/s12868-015-0142-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 02/05/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Testosterone attenuates postnatal hippocampal neurogenesis in adolescent male rhesus macaques through altering neuronal survival. While brain-derived neurotropic factor (BDNF)/ tyrosine kinase receptor B (TrkB) are critical in regulating neuronal survival, it is not known if the molecular mechanism underlying testosterone's action on postnatal neurogenesis involves changes in BDNF/TrkB levels. First, (1) we sought to localize the site of synthesis of the full length and truncated TrkB receptor in the neurogenic regions of the adolescent rhesus macaque hippocampus. Next, (2) we asked if gonadectomy or sex hormone replacement altered hippocampal BDNF and TrkB expression level in mammalian hippocampus (rhesus macaque and Sprague Dawley rat), and (3) if the relationship between BDNF/TrkB expression was altered depending on the sex steroid environment. RESULTS We find that truncated TrkB mRNA+ cells are highly abundant in the proliferative subgranular zone (SGZ) of the primate hippocampus; in addition, there are scant and scattered full length TrkB mRNA+ cells in this region. Gonadectomy or sex steroid replacement did not alter BDNF or TrkB mRNA levels in young adult male rat or rhesus macaque hippocampus. In the monkey and rat, we find a positive correlation with cell proliferation and TrkB-TK+ mRNA expression, and this positive relationship was found only when sex steroids were present. CONCLUSIONS We suggest that testosterone does not down-regulate neurogenesis at adolescence via overall changes in BDNF or TrkB expression. However, BDNF/TrkB mRNA appears to have a greater link to cell proliferation in the presence of circulating testosterone.
Collapse
Affiliation(s)
- Katherine M Allen
- Schizophrenia Research Institute, Sydney, Australia. .,Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Randwick, NSW, 2031, Australia. .,School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Tertia D Purves-Tyson
- Schizophrenia Research Institute, Sydney, Australia. .,Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Randwick, NSW, 2031, Australia. .,School of Medical Sciences, University of New South Wales, Sydney, Australia.
| | - Samantha J Fung
- Schizophrenia Research Institute, Sydney, Australia. .,Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Randwick, NSW, 2031, Australia. .,School of Psychiatry, University of New South Wales, Sydney, Australia.
| | - Cynthia Shannon Weickert
- Schizophrenia Research Institute, Sydney, Australia. .,Schizophrenia Research Laboratory, Neuroscience Research Australia, Barker Street, Randwick, NSW, 2031, Australia. .,School of Psychiatry, University of New South Wales, Sydney, Australia.
| |
Collapse
|
45
|
Abstract
Sex differences in brain function underlie robust differences between males and females in both normal and disease states. Although alternative mechanisms exist, sexual differentiation of the male mammalian brain is initiated predominantly by testosterone secreted by the testes during the perinatal period. Despite considerable advances in understanding how testosterone and its metabolite estradiol sexually differentiate the brain, little is known about the mechanism that generates the male-specific perinatal testosterone surge. In mice, we show that a male-specific activation of GnRH neurons occurs 0-2 h following birth and that this correlates with the male-specific surge of testosterone occurring up to 5 h after birth. The necessity of GnRH signaling for the sexually differentiating effects of the perinatal testosterone surge was demonstrated by the persistence of female-like brain characteristics in adult male, GnRH receptor knock-out mice. Kisspeptin neurons have recently been identified to be potent, direct activators of GnRH neurons. We demonstrate that a population of kisspeptin neurons appears in the preoptic area of only the male between E19 and P1. The importance of kisspeptin inputs to GnRH neurons for the process of sexual differentiation was demonstrated by the lack of a normal neonatal testosterone surge, and disordered brain sexual differentiation of male mice in which the kisspeptin receptor was deleted selectively from GnRH neurons. These observations demonstrate the necessity of perinatal GnRH signaling for driving brain sexual differentiation and indicate that kisspeptin inputs to GnRH neurons are essential for this process to occur.
Collapse
|
46
|
Semaan SJ, Kauffman AS. Daily successive changes in reproductive gene expression and neuronal activation in the brains of pubertal female mice. Mol Cell Endocrinol 2015; 401:84-97. [PMID: 25498961 PMCID: PMC4312730 DOI: 10.1016/j.mce.2014.11.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 10/22/2014] [Accepted: 11/18/2014] [Indexed: 01/01/2023]
Abstract
Puberty is governed by the secretion of gonadotropin releasing hormone (GnRH), but the roles and identities of upstream neuropeptides that control and time puberty remain poorly understood. Indeed, how various reproductive neural gene systems change before and during puberty, and in relation to one another, is not well-characterized. We detailed the daily pubertal profile (from postnatal day [PND] 15 to PND 30) of neural Kiss1 (encoding kisspeptin), Kiss1r (kisspeptin receptor), Tac2 (neurokinin B), and Rfrp (RFRP-3, mammalian GnIH) gene expression and day-to-day c-fos induction in each of these cell types in developing female mice. Kiss1 expression in the AVPV/PeN increased substantially over the pubertal transition, reaching adult levels around vaginal opening (PND 27.5), a pubertal marker. However, AVPV/PeN Kiss1 neurons were not highly activated, as measured by c-fos co-expression, at any pubertal age. In the ARC, Kiss1 and Tac2 cell numbers showed moderate increases across the pubertal period, and neuronal activation of Tac2/Kiss1 cells was moderately elevated at all pubertal ages. Additionally, Kiss1r expression specifically in GnRH neurons was already maximal by PND 15 and did not change with puberty. Conversely, both Rfrp expression and Rfrp/c-fos co-expression in the DMN decreased markedly in the early pre-pubertal stage. This robust decrease of the inhibitory RFRP-3 population may diminish inhibition of GnRH neurons during early puberty. Collectively, our data identify the precise timing of important developmental changes - and in some cases, lack thereof - in gene expression and neuronal activation of key reproductive neuropeptides during puberty, with several changes occurring well before vaginal opening.
Collapse
Affiliation(s)
- Sheila J Semaan
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA
| | - Alexander S Kauffman
- Department of Reproductive Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
47
|
Shirota M, Kawashima J, Nakamura T, Kamiie J, Shirota K, Yoshida M. Dose-dependent acceleration in the delayed effects of neonatal oral exposure to low-dose 17α-ethynylestradiol on reproductive functions in female Sprague-Dawley rats. J Toxicol Sci 2015; 40:727-38. [DOI: 10.2131/jts.40.727] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Mariko Shirota
- Laboratory of Comparative Toxicology, School of Veterinary Medicine
| | - Jun Kawashima
- Laboratory of Comparative Toxicology, School of Veterinary Medicine
| | | | | | - Kinji Shirota
- Laboratory of Veterinary Pathology, Azabu University
- Research Institute of Biosciences, Azabu University
| | - Midori Yoshida
- Division of Pathology, National Institute of Health Sciences
| |
Collapse
|
48
|
Environmental Health Factors and Sexually Dimorphic Differences in Behavioral Disruptions. Curr Environ Health Rep 2014; 1:287-301. [PMID: 25705580 DOI: 10.1007/s40572-014-0027-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mounting evidence suggests that environmental factors-in particular, those that we are exposed to during perinatal life-can dramatically shape the organism's risk for later diseases, including neurobehavioral disorders. However, depending on the environmental insult, one sex may demonstrate greater vulnerability than the other sex. Herein, we focus on two well-defined extrinsic environmental factors that lead to sexually dimorphic behavioral differences in animal models and linkage in human epidemiological studies. These include maternal or psychosocial stress (such as social stress) and exposure to endocrine-disrupting compounds (such as one of the most prevalent, bisphenol A [BPA]). In general, the evidence suggests that early environmental exposures, such as BPA and stress, lead to more pronounced behavioral deficits in males than in females, whereas female neurobehavioral patterns are more vulnerable to later in life stress. These findings highlight the importance of considering sex differences and developmental timing when examining the effects of environmental factors on later neurobehavioral outcomes.
Collapse
|
49
|
Rebuli ME, Cao J, Sluzas E, Delclos KB, Camacho L, Lewis SM, Vanlandingham MM, Patisaul HB. Investigation of the effects of subchronic low dose oral exposure to bisphenol A (BPA) and ethinyl estradiol (EE) on estrogen receptor expression in the juvenile and adult female rat hypothalamus. Toxicol Sci 2014; 140:190-203. [PMID: 24752507 DOI: 10.1093/toxsci/kfu074] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Concerns have been raised regarding the long-term impacts of early life exposure to the ubiquitous environmental contaminant bisphenol A (BPA) on brain organization. Because BPA has been reported to affect estrogen signaling, and steroid hormones play a critical role in brain sexual differentiation, there is also concern that BPA exposure could alter neural sex differences. Here, we examine the impact of subchronic exposure from gestation to adulthood to oral doses of BPA below the current no-observed-adverse-effect level (NOAEL) of 5 mg/kg body weight (bw)/day on estrogen receptor (ESR) expression in sexually dimorphic brain regions of prepubertal and adult female rats. The dams were gavaged daily with vehicle (0.3% carboxymethylcellulose), 2.5, 25, 260, or 2700 μg BPA/kg bw/day, or 0.5 or 5.0 μg ethinyl estradiol (EE)/kg bw/day from gestational day 6 until labor began. Offspring were then gavaged directly from the day after birth until the day before scheduled sacrifice on postnatal days 21 or 90. Using in situ hybridization, one or more BPA doses produced significant decreases in Esr1 expression in the juvenile female rat anteroventral periventricular nucleus (AVPV) of the hypothalamus and significant decreases in Esr2 expression in the adult female rat AVPV and medial preoptic area (MPOA), relative to vehicle controls. BPA did not simply reproduce EE effects, indicating that BPA is not acting solely as an estrogen mimic. The possible consequences of long-term changes in hypothalamic ESR expression resulting from subchronic low dose BPA exposure on neuroendocrine effects are discussed and being addressed in ongoing, related work.
Collapse
Affiliation(s)
- Meghan E Rebuli
- Department of Biology, North Carolina State University, Raleigh, North Carolina 27695 Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Jinyan Cao
- Department of Biology, North Carolina State University, Raleigh, North Carolina 27695 Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - Emily Sluzas
- Department of Biology, North Carolina State University, Raleigh, North Carolina 27695
| | - K Barry Delclos
- National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Luísa Camacho
- National Center for Toxicological Research, Jefferson, Arkansas 72079
| | - Sherry M Lewis
- National Center for Toxicological Research, Jefferson, Arkansas 72079
| | | | - Heather B Patisaul
- Department of Biology, North Carolina State University, Raleigh, North Carolina 27695 Keck Center for Behavioral Biology, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
50
|
Dygalo NN, Shemenkova TV, Kalinina TS, Shishkina GT. A critical point of male gonad development: neuroendocrine correlates of accelerated testicular growth in rats during early life. PLoS One 2014; 9:e93007. [PMID: 24695464 PMCID: PMC3973631 DOI: 10.1371/journal.pone.0093007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/28/2014] [Indexed: 01/08/2023] Open
Abstract
Testis growth during early life is important for future male fertility and shows acceleration during the first months of life in humans. This acceleration coincides with the peak in gonadotropic hormones in the blood, while the role of hypothalamic factors remains vague. Using neonatal rats to assess this issue, we found that day 9 of life is likely critical for testis development in rats. Before this day, testicular growth was proportional to body weight gain, but after that the testes showed accelerated growth. Hypothalamic kisspeptin and its receptor mRNA levels begin to elevate 2 days later, at day 11. A significant increase in the mRNA levels for gonadotropin-releasing hormone (GnRH) receptors in the hypothalamus between days 5 and 7 was followed by a 3-fold decrease in GnRH mRNA levels in this brain region during the next 2 days. Starting from day 9, hypothalamic GnRH mRNA levels increased significantly and positively correlated with accelerated testicular growth. Triptorelin, an agonist of GnRH, at a dose that had no effect on testicular growth during “proportional” period, increased testis weights during the period of accelerated growth. The insensitivity of testicular growth to GnRH during “proportional” period was supported by inability of a 2.5-fold siRNA knockdown of GnRH expression in the hypothalamus of the 7-day-old animals to produce any effect on their testis weights. GnRH receptor blockade with cetrorelix was also without effect on testis weights during “proportional” period but the same doses of this GnRH antagonist significantly inhibited “accelerated” testicular growth. GnRH receptor mRNA levels in the pituitary as well as plasma LH concentrations were higher during “accelerated” period of testicular growth than during “proportional” period. In general, our data defined two distinct periods in rat testicular development that are primarily characterized by different responses to GnRH signaling.
Collapse
Affiliation(s)
- Nikolay N. Dygalo
- Institute of Cytology and Genetics Russian Academy of Sciences, Laboratory of Functional Neurogenomics, Novosibirsk, Russia
- Novosibirsk State University, Department of Physiology, Novosibirsk, Russia
- * E-mail:
| | | | - Tatjana S. Kalinina
- Institute of Cytology and Genetics Russian Academy of Sciences, Laboratory of Functional Neurogenomics, Novosibirsk, Russia
- Novosibirsk State University, Department of Physiology, Novosibirsk, Russia
| | - Galina T. Shishkina
- Institute of Cytology and Genetics Russian Academy of Sciences, Laboratory of Functional Neurogenomics, Novosibirsk, Russia
| |
Collapse
|