1
|
Flannery JC, Tirrell PS, Baumgartner NE, Daniel JM. Neuroestrogens, the hippocampus, and female cognitive aging. Horm Behav 2025; 170:105710. [PMID: 40036999 DOI: 10.1016/j.yhbeh.2025.105710] [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: 08/30/2024] [Revised: 12/30/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025]
Abstract
Research conducted over the last several decades implicates ovarian estrogens as important modulators of hippocampal function. More recently however, the importance of estrogens synthesized in the brain de novo for hippocampal function has been recognized. These brain-derived neuroestrogens act in the hippocampus to regulate dendritic spine dynamics and synaptic plasticity as well as hippocampus-dependent memory. The current report provides an overview of research conducted in model systems elucidating the actions of neuroestrogens in the hippocampus and the subsequent consequences for cognition. We highlight the relationship between ovarian estrogens and brain-derived estrogens and discuss implications for female cognitive aging of the putative decline in hippocampal levels of neuroestrogens following loss of ovarian function. Finally, we propose a model of menopause in which a short-term period of midlife estradiol treatment changes the trajectory of hippocampal neuroestrogen production long-term, resulting in sustained interactions of neuroestrogens, insulin-like growth factor-1, and estrogen receptor signaling in the hippocampus, interactions that support successful brain and cognitive aging.
Collapse
Affiliation(s)
- Jill C Flannery
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Parker S Tirrell
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America
| | - Nina E Baumgartner
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States of America
| | - Jill M Daniel
- Brain Institute, Tulane University, New Orleans, LA 70118, United States of America; Neuroscience Program, Tulane University, New Orleans, LA 70118, United States of America; Department of Psychology, Tulane University, New Orleans, LA, 70118, United States of America.
| |
Collapse
|
2
|
Yarmohammadi-Samani P, Vatanparast J. Sex-specific dendritic morphology of hippocampal pyramidal neurons in the adolescent and young adult rats. Int J Dev Neurosci 2024; 84:47-63. [PMID: 37933732 DOI: 10.1002/jdn.10307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 09/02/2023] [Accepted: 10/16/2023] [Indexed: 11/08/2023] Open
Abstract
CA1 and CA3 pyramidal neurons are the major sources of hippocampal efferents. The structural features of these neurons are presumed to be involved in various normal/abnormal cognitive and emotional outcomes by influencing the pattern of synaptic inputs and neuronal signal processing. Although many studies have described hippocampal structure differences between males and females, these reports mainly focused on gross anatomical features in adult or aged models, and such distinctions on neuronal morphology and dendritic spine density during adolescence, a period of high vulnerability to neurodevelopmental disorders, have received much less attention. In this work, we analyzed dendritic architecture and density of spines in CA1 and CA3 neurons of male and female rats in early adolescence (postnatal day, PND 40) and compared them with those in late adolescence/young adulthood (PND 60). On PND 40, CA1 neurons of male rats showed more Sholl intersections and spine density in apical and basal dendrites compared to those in females. The Sholl intersections in basal dendrites of CA3 neurons were also more in males, whereas the number of apical dendrite intersections was not significantly different between sexes. In male rats, there was a notable decrease in the number of branch and terminal points in the basal dendrite of CA1 neurons of young adults when compared to their sex-matched adolescent rats. On the other hand, CA1 neurons in young adult females also showed more Sholl intersections in apical and basal dendrites compared to adolescent females. Meanwhile, the total cable length, the number of branches, and terminal points of apical dendrites in CA3 neurons also exhibited a significant reduction in young adult male rats compared to their sex-matched adolescents. In young adult rats, both apical and basal dendrites of CA3 neurons in males showed fewer intersections with Sholl circles, but there were no significant differences in dendritic spine density or count estimation between males and females. On the other hand, young adult female rats had more Sholl intersections and dendritic spine count on the basal dendrites of CA3 neurons compared to adolescent females. Although no significant sex- and age-dependent difference in neuronal density was detected in CA1 and CA3 subareas, CA3 pyramidal neurons of both male and female rats showed reduced soma area compared to adolescent rats. Our findings show that the sex differences in the dendritic structure of CA1 and CA3 neurons vary by age and also by the compartments of dendritic arbors. Such variations in the morphology of hippocampal pyramidal neurons may take part as a basis for normal cognitive and affective differences between the sexes, as well as distinct sensitivity to interfering factors and the prevalence of neuropsychological diseases.
Collapse
Affiliation(s)
| | - Jafar Vatanparast
- Department of Biology, School of Science, Shiraz University, Shiraz, Iran
| |
Collapse
|
3
|
Boyd HM, Frick KM, Kwapis JL. Connecting the Dots: Potential Interactions Between Sex Hormones and the Circadian System During Memory Consolidation. J Biol Rhythms 2023; 38:537-555. [PMID: 37464775 PMCID: PMC10615791 DOI: 10.1177/07487304231184761] [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] [Indexed: 07/20/2023]
Abstract
Both the circadian clock and sex hormone signaling can strongly influence brain function, yet little is known about how these 2 powerful modulatory systems might interact during complex neural processes like memory consolidation. Individually, the molecular components and action of each of these systems have been fairly well-characterized, but there is a fundamental lack of information about how these systems cooperate. In the circadian system, clock genes function as timekeeping molecules that convey time-of-day information on a well-stereotyped cycle that is governed by the suprachiasmatic nucleus. Keeping time is particularly important to synchronize various physiological processes across the brain and body, including those that regulate memory consolidation. Similarly, sex hormones are powerful modulators of memory, with androgens, estrogens, and progestins, all influencing memory consolidation within memory-relevant brain regions like the hippocampus. Despite clear evidence that each system can influence memory individually, exactly how the circadian and hormonal systems might interact to impact memory consolidation remains unclear. Research investigating either sex hormone action or circadian gene function within memory-relevant brain regions has unveiled several notable places in which the two systems could interact to control memory. Here, we bring attention to known interactions between the circadian clock and sex hormone signaling. We then review sex hormone-mediated control of memory consolidation, highlighting potential nodes through which the circadian system might interact during memory formation. We suggest that the bidirectional relationship between these two systems is essential for proper control of memory formation based on an animal's hormonal and circadian state.
Collapse
Affiliation(s)
- Hannah M. Boyd
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin
| | - Janine L. Kwapis
- Department of Biology, The Pennsylvania State University, University Park, Pennsylvania
- Center for Eukaryotic Gene Regulation, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
4
|
Phillips-Farfán BV, Quintanar BG, Reyes R, Fernández-Guasti A. Distribution of estrogen receptors alpha and beta in the brain of male rats with same-sex preference. Physiol Behav 2023; 268:114237. [PMID: 37192686 DOI: 10.1016/j.physbeh.2023.114237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/18/2023]
Abstract
Two distinct estrogen receptors (ERs) exist, ERα and ERβ. Both receptors participate in sexual differentiation of the rat brain and likely participate in the regulation of adult sexual orientation (i.e. partner preference). This last idea was investigated herein by examining males treated with the aromatase inhibitor, letrozole, administered prenatally (0.56 μg/kg G10-22). This treatment usually provokes same-sex preference in 1-2 males per litter. Vehicle-treated males (with female preference) and females in spontaneous proestrus (with male preference) were included as controls. ERα and ERβ expression was analyzed by immunohistochemistry in brain areas known to control masculine sexual behavior and partner preference, like the medial preoptic area (MPOA), bed nucleus of the stria terminalis (BNST), medial amygdala (MeA) and ventromedial hypothalamic nucleus (VMH), as well as other brain regions suspected to participate in these processes. In addition, serum levels of estradiol were determined in all male groups. Letrozole-treated male rats that preferred sexually experienced males (LPM) showed over-expressed ERα in the hippocampal cornu Ammonis (CA 1, 3, 4) and dentate gyrus. The LPM group showed up-regulated ERβ expression in the CA2 and reticular thalamic nucleus. The levels of estradiol did not differ between the groups. Higher expression of ERs in these males was different than their expression in females, with male sex-preference. This suggests that males with same-sex preference showed a unique brain, this sui generis steroid receptor expression probably participates in the biological underpinnings of sexual preference.
Collapse
Affiliation(s)
| | | | - Rebeca Reyes
- Departament of Pharmacobiology, Cinvestav, Unidad Coapa, Mexico City, Mexico
| | | |
Collapse
|
5
|
Lupu DI, Cediel Ulloa A, Rüegg J. Endocrine-Disrupting Chemicals and Hippocampal Development: The Role of Estrogen and Androgen Signaling. Neuroendocrinology 2023; 113:1193-1214. [PMID: 37356425 DOI: 10.1159/000531669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 06/06/2023] [Indexed: 06/27/2023]
Abstract
Hormones are important regulators of key processes during fetal brain development. Thus, the developing brain is vulnerable to the action of chemicals that can interfere with endocrine signals. Epidemiological studies have pointed toward sexually dimorphic associations between neurodevelopmental outcomes, such as cognitive abilities, in children and prenatal exposure to endocrine-disrupting chemicals (EDCs). This points toward disruption of sex steroid signaling in the development of neural structures underlying cognitive functions, such as the hippocampus, an essential mediator of learning and memory processes. Indeed, during development, the hippocampus is subjected to the organizational effects of estrogens and androgens, which influence hippocampal cell proliferation, differentiation, dendritic growth, and synaptogenesis in the hippocampal fields of Cornu Ammonis and the dentate gyrus. These early organizational effects correlate with a sexual dimorphism in spatial cognition and are subject to exogenous chemical perturbations. This review summarizes the current knowledge about the organizational effects of estrogens and androgens on the developing hippocampus and the evidence for hippocampal-dependent learning and memory perturbations induced by developmental exposure to EDCs. We conclude that, while it is clear that sex hormone signaling plays a significant role during hippocampal development, a complete picture at the molecular and cellular levels would be needed to establish causative links between the endocrine modes of action exerted by EDCs and the adverse outcomes these chemicals can induce at the organism level.
Collapse
Affiliation(s)
- Diana-Ioana Lupu
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | | | - Joëlle Rüegg
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
LaDage LD. Seasonal variation in gonadal hormones, spatial cognition, and hippocampal attributes: More questions than answers. Horm Behav 2022; 141:105151. [PMID: 35299119 DOI: 10.1016/j.yhbeh.2022.105151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/04/2022]
Abstract
A large body of research has been dedicated to understanding the factors that modulate spatial cognition and attributes of the hippocampus, a highly plastic brain region that underlies spatial processing abilities. Variation in gonadal hormones impacts spatial memory and hippocampal attributes in vertebrates, although the direction of the effect has not been entirely consistent. To add complexity, individuals in the field must optimize fitness by coordinating activities with the appropriate environmental cues, and many of these behaviors are correlated tightly with seasonal variation in gonadal hormone release. As such, it remains unclear if the relationship among systemic gonadal hormones, spatial cognition, and the hippocampus also exhibits seasonal variation. This review presents an overview of the relationship among gonadal hormones, the hippocampus, and spatial cognition, and how the seasonal release of gonadal hormones correlates with seasonal variation in spatial cognition and hippocampal attributes. Additionally, this review presents other neuroendocrine mechanisms that may be involved in modulating the relationship among seasonality, gonadal hormone release, and the hippocampus and spatial cognition, including seasonal rhythms of steroid hormone binding globulins, neurosteroids, sex steroid hormone receptor expression, and hormone interactions. Here, endocrinology, ecology, and behavioral neuroscience are brought together to present an overview of the research demonstrating the mechanistic effects of systemic gonadal hormones on spatial cognition and the hippocampus, while, at a functional level, superimposing seasonal effects to examine ecologically-relevant circannual changes in gonadal hormones and spatial behaviors.
Collapse
Affiliation(s)
- Lara D LaDage
- Penn State Altoona, Division of Mathematics & Natural Sciences, 3000 Ivyside Dr., Altoona, PA 16601, USA.
| |
Collapse
|
7
|
Lichtensteiger W, Bassetti-Gaille C, Rehrauer H, Georgijevic JK, Tresguerres JAF, Schlumpf M. Converging Effects of Three Different Endocrine Disrupters on Sox and Pou Gene Expression in Developing Rat Hippocampus: Possible Role of microRNA in Sex Differences. Front Genet 2021; 12:718796. [PMID: 34858468 PMCID: PMC8632217 DOI: 10.3389/fgene.2021.718796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) can impair hippocampus-dependent behaviors in rat offspring and in children. In search for key processes underlying this effect, we compared the transcriptomes of rat hippocampus on postnatal day 6 after gestational and lactational exposure to three different EDCs at doses known to impair development of learning and memory. Aroclor 1254, a commercial PCB mixture (5 mg/kg or 0.5 mg/kg), or bisphenol A (5 mg/kg or 0.5 mg/kg) were administered in chow, chlorpyrifos (3 mg/kg or 1 mg/kg) was injected subcutaneously. Male hippocampus exhibited a common effect of all three chemicals on genes involved in cell-autonomous processes, Sox6, Sox11, Pou2f2/Oct2, and Pou3f2/Brn2, all upregulated at the high dose. Additional genes of the Sox and Pou families were affected by only one or two of the chemicals. Real time RT PCR showed a comparable expression change for bisphenol A also at the lower dose. Female hippocampus exhibited much fewer genes with expression changes (almost none with false discovery rate <0.05), and none of the genes of the Sox and Pou families was affected. Since gene network analyses in male hippocampus suggested a link between Sox6 and miR-24, known to be repressed by activation of ER-alpha and to repress Sox6 in other tissues, this microRNA was measured. miR-24 was downregulated by all chemicals at the high dose in males. Values of Sox6 mRNA and miR-24 were inversely correlated in individual male hippocampus samples, supporting the hypothesis that the change in Sox6 expression resulted from an action of miR-24. In contrast, miR-24 levels remained unchanged in hippocampus of females. A sexually dimorphic response of miR-24 may thus be at the basis of the sex difference in Sox6 expression changes following exposure to the three chemicals. ER-alpha expression was also sex-dependent, but the expression changes did not parallel those of potential downstream genes such as Sox6. Sox6 is known to suppress differentiation of Parvalbumin (Pvalb)-expressing interneurons. Individual Sox6 levels (FPKM) were inversely correlated with levels of Pvalb, but not with markers of Sox6-independent interneuron subpopulations, Nos1 and 5HT3aR. Effects on interneuron development are further suggested, in males, by expression changes of Nrg1 and its receptor Erbb4, controlling interneuron migration. Our study disclosed new types of EDC-responsive morphogenetic genes, and illustrated the potential relevance of microRNAs in sexually dimorphic EDC actions.
Collapse
Affiliation(s)
- Walter Lichtensteiger
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Catherine Bassetti-Gaille
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | - Jelena Kühn Georgijevic
- Functional Genomics Center, Swiss Federal Institute of Technology and University of Zurich, Zurich, Switzerland
| | | | - Margret Schlumpf
- GREEN Tox and Institute of Veterinary Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
8
|
Brann DW, Lu Y, Wang J, Zhang Q, Thakkar R, Sareddy GR, Pratap UP, Tekmal RR, Vadlamudi RK. Brain-derived estrogen and neural function. Neurosci Biobehav Rev 2021; 132:793-817. [PMID: 34823913 PMCID: PMC8816863 DOI: 10.1016/j.neubiorev.2021.11.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/26/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023]
Abstract
Although classically known as an endocrine signal produced by the ovary, 17β-estradiol (E2) is also a neurosteroid produced in neurons and astrocytes in the brain of many different species. In this review, we provide a comprehensive overview of the localization, regulation, sex differences, and physiological/pathological roles of brain-derived E2 (BDE2). Much of what we know regarding the functional roles of BDE2 has come from studies using specific inhibitors of the E2 synthesis enzyme, aromatase, as well as the recent development of conditional forebrain neuron-specific and astrocyte-specific aromatase knockout mouse models. The evidence from these studies support a critical role for neuron-derived E2 (NDE2) in the regulation of synaptic plasticity, memory, socio-sexual behavior, sexual differentiation, reproduction, injury-induced reactive gliosis, and neuroprotection. Furthermore, we review evidence that astrocyte-derived E2 (ADE2) is induced following brain injury/ischemia, and plays a key role in reactive gliosis, neuroprotection, and cognitive preservation. Finally, we conclude by discussing the key controversies and challenges in this area, as well as potential future directions for the field.
Collapse
Affiliation(s)
- Darrell W Brann
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Jing Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Roshni Thakkar
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Gangadhara R Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Uday P Pratap
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Rajeshwar R Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health, San Antoio TX, 78229, USA; Audie L. Murphy Division, South Texas Veterans Health Care System, San Antonio, TX, 78229, USA.
| |
Collapse
|
9
|
Qian L, Qi S, Zhang J, Duan M, Schlenk D, Jiang J, Wang C. Exposure to Boscalid Induces Reproductive Toxicity of Zebrafish by Gender-Specific Alterations in Steroidogenesis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2020; 54:14275-14287. [PMID: 33138376 DOI: 10.1021/acs.est.0c02871] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Boscalid is a succinate dehydrogenase inhibitor fungicide and is frequently detected in surface water. Due to the frequent detection of boscalid, we evaluated its impact on the reproduction of adult zebrafish following a 21 d exposure to 0, 0.01, 0.1, and 1.0 mg/L. Following exposure to boscalid, the fertility of female zebrafish and fertilization rate of spawning eggs were reduced in a concentration-dependent manner up to a respective 87% and 20% in the highest concentration. A significant 16% reduction in the percentage of late vitellogenic oocytes was noted in ovaries, and a significant 74% reduction in the percentage of spermatids in testis was also observed after treatment with 1.0 mg/L. 17β-Estradiol (E2) concentrations decreased significantly in females (34% decrease) but significantly increased in males (15% increase) following 1.0 mg/L boscalid treatment. The expression of genes (such as era, er2b, cyp19a, and cyp19b) related to the hypothalamus-pituitary-gonad-liver (HPGL) axis was significantly altered and positively correlated with E2 concentrations in female and male zebrafish (p < 0.05). Molecular docking results revealed that the binding modes between boscalid and target proteins (ER and CYP19) of zebrafish were similar to that of the reference compounds and the target proteins. The binding energies indicate that boscalid may have a weak estrogen-like binding effect or CYP19 inhibition, potentially altering the HPGL axis, thereby reducing E2 concentrations and fecundity in females. In contrast, boscalid caused significant induction of E2 steroidogenesis and subsequent feminization of gonads in males, indicating gender-specific adverse outcome pathways.
Collapse
Affiliation(s)
- Le Qian
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Suzhen Qi
- Risk Assessment Laboratory for Bee Product Quality and Safety of Ministry of Agriculture, Institute of Agricultural Research, Chinese Academy of Agricultural Sciences, Beijing 100093, People's Republic of China
| | - Jie Zhang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Manman Duan
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Daniel Schlenk
- Department of Environmental Sciences, University of California, Riverside, Riverside, California 92521, United States
| | - Jiazhen Jiang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| | - Chengju Wang
- College of Sciences, China Agricultural University, Beijing 100193, People's Republic of China
| |
Collapse
|
10
|
Premachandran H, Zhao M, Arruda-Carvalho M. Sex Differences in the Development of the Rodent Corticolimbic System. Front Neurosci 2020; 14:583477. [PMID: 33100964 PMCID: PMC7554619 DOI: 10.3389/fnins.2020.583477] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, a growing body of research has shown sex differences in the prevalence and symptomatology of psychopathologies, such as depression, anxiety, and fear-related disorders, all of which show high incidence rates in early life. This has highlighted the importance of including female subjects in animal studies, as well as delineating sex differences in neural processing across development. Of particular interest is the corticolimbic system, comprising the hippocampus, amygdala, and medial prefrontal cortex. In rodents, these corticolimbic regions undergo dynamic changes in early life, and disruption to their normative development is believed to underlie the age and sex-dependent effects of stress on affective processing. In this review, we consolidate research on sex differences in the hippocampus, amygdala, and medial prefrontal cortex across early development. First, we briefly introduce current principles on sexual differentiation of the rodent brain. We then showcase corticolimbic regional sex differences in volume, morphology, synaptic organization, cell proliferation, microglia, and GABAergic signaling, and explain how these differences are influenced by perinatal and pubertal gonadal hormones. In compiling this research, we outline evidence of what and when sex differences emerge in the developing corticolimbic system, and illustrate how temporal dynamics of its maturational trajectory may differ in male and female rodents. This will help provide insight into potential neural mechanisms underlying sex-specific critical windows for stress susceptibility and behavioral emergence.
Collapse
Affiliation(s)
| | - Mudi Zhao
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
11
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
12
|
Abstract
The hippocampus is central to spatial learning and stress responsiveness, both of which differ in form and function in males versus females, yet precisely how the hippocampus contributes to these sex differences is largely unknown. In reproductively mature individuals, sex differences in the steroid hormone milieu undergirds many sex differences in hippocampal-related endpoints. However, there is also evidence for developmental programming of adult hippocampal function, with a central role for androgens as well as their aromatized byproduct, estrogens. These include sex differences in cell genesis, synapse formation, dendritic arborization, and excitatory/inhibitory balance. Enduring effects of steroid hormone modulation occur during two developmental epochs, the first being the classic perinatal critical period of sexual differentiation of the brain and the other being adolescence and the associated hormonal changes of puberty. The cellular mechanisms by which steroid hormones enduringly modify hippocampal form and function are poorly understood, but we here review what is known and highlight where attention should be focused.
Collapse
|
13
|
Acaz-Fonseca E, Castelló-Ruiz M, Burguete MC, Aliena-Valero A, Salom JB, Torregrosa G, García-Segura LM. Insight into the molecular sex dimorphism of ischaemic stroke in rat cerebral cortex: Focus on neuroglobin, sex steroids and autophagy. Eur J Neurosci 2020; 52:2756-2770. [PMID: 32243028 DOI: 10.1111/ejn.14731] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/12/2020] [Accepted: 03/22/2020] [Indexed: 12/12/2022]
Abstract
Including sex is of paramount importance in preclinical and clinical stroke researches, and molecular studies dealing in depth with sex differences in stroke pathophysiology are needed. To gain insight into the molecular sex dimorphism of ischaemic stroke in rat cerebral cortex, male and female adult rats were subjected to transient middle cerebral artery occlusion. The expression of neuroglobin (Ngb) and other functionally related molecules involved in sex steroid signalling (oestrogen and androgen receptors), steroidogenesis (StAR, TSPO and aromatase) and autophagic activity (LC3B-II/LC3B-I ratio, UCP2 and HIF-1α) was assessed in the ipsilateral ischaemic and contralateral non-ischaemic hemispheres. An increased expression of Ngb was detected in the injured female cerebral cortex. In contrast, increased expression of oestrogen receptor α, GPER, StAR, TSPO and UCP2, and decreased androgen receptor expression were detected in the injured male cortex. In both sexes, the ischaemic insult induced an upregulation of LC3B-II/-I ratio, indicative of increased autophagy. Therefore, the cerebral cortex activates both sex-specific and common molecular responses with neuroprotective potential after ischaemia-reperfusion, which globally results in similar stroke outcome in both sexes. Nonetheless, these different potential molecular targets should be taken into account when neuroprotective drugs aiming to reduce brain damage in ischaemic stroke are investigated.
Collapse
Affiliation(s)
- Estefanía Acaz-Fonseca
- Instituto Cajal - CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Castelló-Ruiz
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - Alicia Aliena-Valero
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | - Juan B Salom
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - Germán Torregrosa
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe - Universidad de Valencia, Valencia, Spain
| | - Luis M García-Segura
- Instituto Cajal - CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Finney CA, Shvetcov A, Westbrook RF, Jones NM, Morris MJ. The role of hippocampal estradiol in synaptic plasticity and memory: A systematic review. Front Neuroendocrinol 2020; 56:100818. [PMID: 31843506 DOI: 10.1016/j.yfrne.2019.100818] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/29/2019] [Accepted: 12/11/2019] [Indexed: 12/31/2022]
Abstract
The consolidation of long-term memory is influenced by various neuromodulators. One of these is estradiol, a steroid hormone that is synthesized both in peripheral endocrine tissue and in the brain, including the hippocampus. Here, we examine the evidence regarding the role of estradiol in the hippocampus, specifically, in memory formation and its effects on the molecular mechanisms underlying synaptic plasticity. We conclude that estradiol improves memory consolidation and, thereby, long-term memory. Previous studies have shown that it does this in three, interconnected ways: (1) via functional changes in excitatory activity, (2) signaling changes in calcium dynamics, protein phosphorylation and protein expression, and (3) structural changes to synaptic morphology. Through a functional network analysis of proteins affected by estradiol, we identify potential protein-protein interactions that further support a role for estradiol in modulating synaptic plasticity as well as highlight signaling pathways that may be involved in these changes within the hippocampus.
Collapse
Affiliation(s)
- C A Finney
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - A Shvetcov
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - R F Westbrook
- School of Psychology, University of New South Wales, Sydney, NSW, Australia
| | - N M Jones
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - M J Morris
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
15
|
Tsai HW, Franklin M, Armoskus C, Taniguchi S, Moder C, Trang K, Santacruz M, Milla A. Androgenic regulation of sexually dimorphic expression of RNA binding motif protein 48 in the developing mouse cortex and hippocampus. Int J Dev Neurosci 2019; 78:33-44. [PMID: 31400491 PMCID: PMC6897302 DOI: 10.1016/j.ijdevneu.2019.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 11/28/2022] Open
Abstract
To further reveal the molecular mechanism underlying sexual differentiation of the mouse cerebral cortex and hippocampus, we reanalyzed our previous microarray study with Gene Ontology (GO) term enrichment and found that the GO term "RNA binding" was over-represented among the 89 sexually dimorphic candidate genes. Thus, we selected 16 autosomal genes annotated to the term RNA binding and profiled their mRNA expression in the developing male and female mouse cortex/hippocampus. During the first three weeks after birth, sex differences in mRNA levels of Khdrbs2, Nanos2, Rbm48, and Tdrd3 were observed in the mouse cortex/hippocampus. Of these genes, only the female-biased expression of Rbm48 in neonates was abolished by prenatal exposure to testosterone propionate (TP), while postnatal treatment of TP three weeks after birth increased Rbm48 and Tdrd3 mRNA levels in both sexes. Regardless of sex, the postnatal cortex/hippocampus also showed a marked increase in the content of androgen receptor (Ar) and estrogen receptor β (Esr2), but a decrease in estrogen receptor α (Esr1) and aromatase (Cyp19a1), which might confer the different responses of Rbm48 to prenatal and postnatal TP. Our results suggest that androgen-regulated, sexually dimorphic Rbm48 expression might present a novel molecular mechanism by which perinatal androgens control development of sexual dimorphism in cortical and hippocampal structure and function.
Collapse
Affiliation(s)
- Houng-Wei Tsai
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Michael Franklin
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Chris Armoskus
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Saori Taniguchi
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Courtney Moder
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Kathy Trang
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Marilisa Santacruz
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Allyson Milla
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| |
Collapse
|
16
|
Green MR, Zeidan M, Hodges TE, McCormick CM. Age-dependent regulation by androgens of gene expression in the anterior hypothalamus and stress-induced release of adrenal hormones in adolescent and adult male rats. J Neuroendocrinol 2019; 31:e12714. [PMID: 30912177 DOI: 10.1111/jne.12714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/06/2019] [Accepted: 03/21/2019] [Indexed: 01/05/2023]
Abstract
Adolescents show greater and/or more prolonged activation of the hypothalamic-pituitary-adrenal axis in response to stressors than adults, although the basis for such an age difference is not understood. We investigated developmental shifts in the regulation of HPA function by testosterone using androgen replacement in orchiectomised (OCX) pre-pubertal and post-pubertal adolescent rats and in adults, as well as using inhibitors of testosterone synthesis in non-operated rats. The expected dampening effect of testosterone in adult OCX rats did not meet statistical significance in all of the three experiments. Nevertheless, in each, adolescents had higher post-stress concentrations of corticosterone compared to adults despite similar concentrations of testosterone. The effect of testosterone was in the opposite direction in post-pubertal adolescents compared to that in adults, with testosterone replacement leading to increased rather than lower corticosterone concentration. Testosterone replacement decreased arginine vasopressin and corticotrophin-releasing hormone immune-reactive cell counts in the paraventricular nucleus at all ages. In a fourth experiment, we provide evidence that the basis of the age difference in corticosterone release is because of a greater conversion of testosterone to oestradiol in adolescents and a greater conversion of testosterone to dihydrotestosterone in adults: aromatase inhibition had little effect in adults and attenuated the age difference by decreasing stress-induced corticosterone release in adolescents. By contrast, 5α-reductase inhibition or an androgen receptor antagonist had little effect in adolescents and attenuated the age difference by increasing stress-induced corticosterone release in adults. In the adrenal gland, adolescents had reduced 5α-reductase and androgen receptor gene expression. There also were age differences in the regulation of hypothalamic mRNA expression of androgen receptors, oestrogen receptors and aromatase by testosterone. In sum, the results suggest that developmental shifts in the synthesis of testosterone and the regulation of gene expression are factors with respect to age differences in corticosterone release in response to stressors.
Collapse
Affiliation(s)
- Matthew R Green
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Mostafa Zeidan
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Travis E Hodges
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Cheryl M McCormick
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
17
|
Arias-Reyes C, Losantos-Ramos K, Gonzales M, Furrer D, Soliz J. NADH-linked mitochondrial respiration in the developing mouse brain is sex-, age- and tissue-dependent. Respir Physiol Neurobiol 2019; 266:156-162. [PMID: 31128272 DOI: 10.1016/j.resp.2019.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/09/2019] [Accepted: 05/20/2019] [Indexed: 11/17/2022]
Abstract
Mitochondria play a major role in the brain. Apart from energy production, mitochondria regulate key factors in the activation of cell signaling pathways such as survival, proliferation, and differentiation. While all these processes occur during the physiological development of the brain, it is surprising that the mitochondrial functions and functioning in the brain during the postnatal development remain poorly explored. In this work, we collected samples of brainstem and cortex of mice at postnatal ages 3 (P3), 21 (P21), and at adulthood (3 months old) and evaluated the mitochondrial oxygen consumption after complex I activation. To do so, we used our oxygraph-2 K system (OROBOROS) that measures the mitochondrial bioenergetics in saponin-permeabilized tissue punches of 2 mg weight. Furthermore, as sex dimorphism in the brain occurs since very early stages of development, we performed experiments in brain samples of male and female mice. Accordingly, the mitochondrial oxygen consumption rate (OCR) was evaluated under activation of complex I (NADH-linked respiration - mitochondrial state 3), and during the inhibition of the complex V (ATP synthase) with oligomycin (mitochondrial state 4). In following, the respiratory control ratio (RCR - state 3/state4) was calculated as an index of mitochondrial oxidative-phosphorylation coupling. Our results show that the activity of the mitochondrial complex I in the brain increases along with the postnatal development in a sex- and tissue-dependent manner, with males showing higher activity than females, and with brainstem tissue showing higher activity than cortex. Our data may contribute to a better understanding of the sex-dependent maturation of the cortex and the cardiorespiratory network located in the brainstem.
Collapse
Affiliation(s)
- C Arias-Reyes
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada; Instituto de Biología Molecular y Biotecnología, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - K Losantos-Ramos
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - M Gonzales
- Instituto Boliviano de Biología de la Altura, Facultad de Medicina, Universidad Mayor de San Andrés, La Paz, Bolivia
| | - D Furrer
- Oncology Axis, CHU of Quebec Research Center, Laval University, Quebec City, Canada
| | - J Soliz
- Institut universitaire de cardiologie et de pneumologie de Québec, Centre Hospitalier Universitaire de Québec (CHUQ), Faculty of Medicine, Université Laval, Québec, QC, Canada; Instituto de Biología Molecular y Biotecnología, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andrés, La Paz, Bolivia.
| |
Collapse
|
18
|
Koss WA, Frick KM. Activation of androgen receptors protects intact male mice from memory impairments caused by aromatase inhibition. Horm Behav 2019; 111:96-104. [PMID: 30653980 PMCID: PMC6527464 DOI: 10.1016/j.yhbeh.2019.01.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022]
Abstract
Although 17β-estradiol (E2) is known to regulate hippocampal function, the specific contributions of hippocampally-synthesized E2 remain unclear. Infusion of the aromatase inhibitor letrozole into the dorsal hippocampus (DH) of ovariectomized mice disrupts object recognition and object placement memory consolidation, suggesting that DH-synthesized E2 is essential for memory. However, the role of DH-synthesized E2 in memory among male rodents is unknown. Here, we examined effects of aromatase inhibition on memory consolidation in male mice. Intact and gonadectomized mice were infused with vehicle or letrozole into the DH immediately post-training in object placement and object recognition tasks. Letrozole blocked memory in both tasks among gonadectomized males only, suggesting that circulating androgens, or a rise in hippocampal androgens due to aromatase inhibition, may support memory consolidation in intact males. To test this hypothesis, intact males were infused with the androgen receptor antagonist flutamide into the DH after object training. A dose-dependent impairment was observed in both tasks, indicating that blocking androgen signaling can impair memory consolidation. To test if hippocampal androgen receptor activation protected intact males from the impairing effects of letrozole, a non-impairing dose of flutamide was co-infused with letrozole. Co-administration of both drugs blocked object placement and object recognition memory consolidation, demonstrating that letrozole impairs memory in intact males only if androgen receptors are blocked. Together, these data suggest that DH-synthesized E2 and androgen receptor activation may work in concert to mediate memory consolidation in intact males, such that androgen receptor activation protects against memory impairments caused by aromatase inhibition.
Collapse
Affiliation(s)
- Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America.
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States of America.
| |
Collapse
|
19
|
Zhang L, Cao LL, Yang DD, Ding JH, Guo XD, Xue TF, Zhao XJ, Sun XL. Establishment and evaluation of a novel mouse model of peri/postmenopausal depression. Heliyon 2019; 5:e01195. [PMID: 30839939 PMCID: PMC6365542 DOI: 10.1016/j.heliyon.2019.e01195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/20/2018] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Women are believed to be more vulnerable to develop depressive symptoms during the perimenopause compared to postmenopause. The traditional bilateral ovariectomy and chronic mild stress (CMS) stimulation animal model produces a postmenopausal depressive-like state but the transition from perimenopausal period to postmenopausal period was ignored. Thus we establish a novel animal model in which the mice were stimulated by CMS for three months and removed the ovaries by two-step operation, and then evaluate whether this novel model could be much better for preclinical study used as a peri/postmenopause depressive model. The present study systemically evaluated the changes induced by two-step ovariectomy plus CMS in the mice. The depression-like behaviors, the levels of corticosterone, estrogen, pro-inflammatory factors, neurotransmitters, as well as brain-derived neurotrophic factor were determined; the changes of estrogen receptors, serotonin receptors, uterine weight and bone microarchitecture were also observed. The results show that the behaviors and biochemical indexes of mice changed gradually over time. Our study suggests that this two-step ovariectomy operation plus CMS successfully establishes a more reasonable peri/postmenopausal depression animal model which effectively simulates the clinical symptoms of peri/postmenopausal depressive women.
Collapse
Affiliation(s)
- Ling Zhang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Lu-Lu Cao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dan-Dan Yang
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jian-Hua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xu-Dong Guo
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Teng-Fei Xue
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiao-Jie Zhao
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiu-Lan Sun
- Neuroprotective Drug Discovery Key Laboratory of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.,Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| |
Collapse
|
20
|
Souldouzi R, Razi M, Shalizar Jalali A, Jalilzadeh-Amin G, Amani S. Effect of (R)-(+) Pulegone on Ovarian Tissue; Correlation with Expression of Aromatase Cyp19 and Ovarian Selected Genes in Mice. CELL JOURNAL 2018; 20:231-243. [PMID: 29633601 PMCID: PMC5893295 DOI: 10.22074/cellj.2018.4798] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 03/03/2017] [Indexed: 12/26/2022]
Abstract
Objective Pulegone (PGN) is a monoterpene ketone, whose metabolites exert several cytotoxic effects in
various tissues. The present study was conducted in order to evaluate the (R)-(+) PGN-induced alterations in
ovarian aromatization, proto-oncogenes and estrogen receptorα (ERα) and ERβ receptors expressions.
Materials and Methods In this experimental study, mature albino mice were divided into experimental (received
25 mg/kg, 50 mg/kg and 100 mg/kg PGN, orally for 35 days) and control (received 2% solution of Tween 80
as a PGN solvent, orally) groups. The mRNA levels of Erα, Erβ, p53, Bcl-2, and cytochrome p450 (Cyp19)
as well as ovarian angiogenesis were analyzed through reverse transcription polymerase chain reaction and
immunohistochemical techniques, respectively. Moreover, apoptosis of follicular cells, serum estrogen and
progesterone levels and mRNA damage were investigated via using terminal transferase and biotin-16-dUTP
staining, electrochemilunescence and fluorescent microscopy methods, respectively.
Results The PGN reduced Erα, Erβ and Cyp19 expression at 50 mg/kg and 100 mg/kg doses, while significantly
elevating p53 and reducing Bcl-2 expression. Finally, PGN impaired ovarian angiogenesis, increased apoptosis,
elevated follicular atresia and reduced serum levels of estrogen and progesterone.
Conclusion Chronic exposure to PGN (50 mg/kg and 100 mg/kg), severely affects ovarian aromatization, proto-
oncogenes mRNA levels and expression of ERs.
Collapse
Affiliation(s)
- Rohiyeh Souldouzi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Mazdak Razi
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Ali Shalizar Jalali
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran.
| | - Ghader Jalilzadeh-Amin
- Department of Veterinary Internal Medicine, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Saeedeh Amani
- Department of Basic Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
21
|
Kight KE, McCarthy MM. Sex differences and estrogen regulation of BDNF gene expression, but not propeptide content, in the developing hippocampus. J Neurosci Res 2017; 95:345-354. [PMID: 27870444 DOI: 10.1002/jnr.23920] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/10/2016] [Accepted: 08/22/2016] [Indexed: 01/29/2023]
Abstract
Sex differences in adult brain function are frequently determined developmentally through the actions of steroid hormones during sensitive periods of prenatal and early postnatal life. In rodents, various cellular end points of the developing brain are affected by estradiol that is derived from the aromatization of circulating testosterone and/or synthesized within the brain. We have previously described a sex difference in neurogenesis in the hippocampus of neonatal rats that is modulated by estradiol. In this report, we examined a potential downstream regulator of the effects of estradiol on hippocampal cell proliferation by measuring gene expression of brain-derived neurotrophin (BDNF) in male and female neonatal rats in response to estradiol. Males had higher baseline BDNF gene expression in dentate gyrus and CA1 regions of the hippocampus compared with females. Neonatal administration of exogenous estradiol resulted in opposite effects on BDNF expression in these areas of the neonatal hippocampus, such that BDNF transcripts increased in CA1 but decreased in dentate. Blocking endogenous estradiol signaling by antagonizing estrogen receptors decreased BDNF expression in the dentate of males, but not females, and had no effect in CA1. Interestingly, this sex difference and response to estradiol was not mirrored by translational output, as no differences in BDNF precursor peptide were observed. The sex- and region-specific effects of estradiol on BDNF expression in the neonatal hippocampus suggest a complex functional relationship between these pleiotropic factors in regulating developmental neurogenesis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katherine E Kight
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Margaret M McCarthy
- Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, Maryland.,Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
22
|
Zhao Y, He L, Zhang Y, Zhao J, Liu Z, Xing F, Liu M, Feng Z, Li W, Zhang J. Estrogen receptor alpha and beta regulate actin polymerization and spatial memory through an SRC-1/mTORC2-dependent pathway in the hippocampus of female mice. J Steroid Biochem Mol Biol 2017; 174:96-113. [PMID: 28789972 DOI: 10.1016/j.jsbmb.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 07/29/2017] [Accepted: 08/03/2017] [Indexed: 12/18/2022]
Abstract
Aging-related decline of estrogens, especially 17β-estradiol (E2), has been shown to play an important role in the impairment of learning and memory in dementias, such as Alzheimer's disease (AD), but the underlying molecular mechanisms are poorly understood. In this study, we first demonstrated decreases in E2 signaling (aromatase, classic estrogen receptor ERα and ERβ and their coactivator SRC-1), mTORC2 signaling (Rictor and phospho-AKTser473) and actin polymerization (phospho-Cofilin, Profilin-1 and the F-actin/G-actin ratio) in the hippocampus of old female mice compared with those levels detected in the adult hippocampus. We then showed that ERα and ERβ antagonists induced a significant decrease in SRC-1, mTORC2 signaling, actin polymerization, and CA1 spine density, as well as impairments of learning and memory; however, ovariectomy-induced changes of these parameters could be significantly reversed by treatment with ER agonists. We further showed that expression of SRC-1, mTORC2 signaling and actin polymerization could be upregulated by E2 treatment, and the effects of E2 were blocked by the ER antagonists but mimicked by the agonists. We also showed that the lentivirus-mediated SRC-1 knockdown significantly inhibited the agonist-activated mTORC2 signaling and actin polymerization, and the lentivirus-mediated Rictor knockdown also significantly inhibited the agonist-activated actin polymerization. Finally, we demonstrated that the ERα and ERβ antagonists induced a disruption in actin polymerization and an impairment of spatial memory, which were rescued by activation of mTORC2. Taken together, the above results clearly demonstrated an mTORC2-dependent regulation of actin polymerization that contributed to the effects of ERα and ERβ on spatial learning, which may provide a novel target for the prevention and treatment of E2-related dementia in the aged population.
Collapse
Affiliation(s)
- Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jikai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Zhi Liu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Fangzhou Xing
- Key Laboratory of Freshwater Fish Reproduction and Development (Ministry of Education), Key Laboratory of Aquatic Science of Chongqing, Key Laboratory of Eco-Environments in Three Gorges Reservoir Region, School of Life Sciences, Southwest University, Chongqing, 400715, China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Ziqi Feng
- Cadet Brigade, Third Military Medical University, Chongqing 400038, China
| | - Wei Li
- School of Nursing, Third Military Medical University, Chongqing 400038, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
23
|
Nelson LH, Warden S, Lenz KM. Sex differences in microglial phagocytosis in the neonatal hippocampus. Brain Behav Immun 2017; 64:11-22. [PMID: 28341582 PMCID: PMC5512447 DOI: 10.1016/j.bbi.2017.03.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/07/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
Microglia regulate brain development through many processes, such as promoting neurogenesis, supporting cell survival, and phagocytizing progenitor, newly-born, and dying cells. Many of these same developmental processes show robust sex differences, yet very few studies have assessed sex differences in microglia function during development. Hormonally-induced sexual differentiation of the brain occurs during the perinatal period, thus we examined sex differences in microglial morphology, phagocytosis, and proliferation in the hippocampus during the early postnatal period. We found that the neonatal female hippocampus had significantly more microglia with phagocytic cups than the male hippocampus. We subsequently found that female microglia phagocytized more neural progenitor cells and healthy cells compared to males, but there were no sex differences in the number of newly-born or dying cells targeted by microglial phagocytosis. We found that the number of phagocytic microglia in females was reduced to male-typical levels by treatment with estradiol, the hormone responsible for masculinizing the rodent brain. Females also had higher expression of several phagocytic pathway genes in the hippocampus compared to males. In contrast to robust sex differences in phagocytic microglia, we found no sex differences in the number of microglia with amoeboid, transitioning, or ramified morphologies or differences in three-dimensional reconstructions of microglial morphology. While we did not find a baseline sex difference in microglial proliferation during or following the prenatal gonadal hormone surge in males, we found that estradiol treatment increased microglia proliferation in females. Overall, these data show that there are important sex differences in microglia function in the hippocampus during the early neonatal period.
Collapse
Affiliation(s)
- Lars H Nelson
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave., Columbus, OH 43210, USA; Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH, USA.
| | - Spencer Warden
- Department of Psychology, The Ohio State University, 1835 Neil Ave, Columbus, OH 43210,Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH
| | - Kathryn M Lenz
- Department of Neuroscience, The Ohio State University, 333 W. 10th Ave., Columbus, OH 43210, USA; Department of Psychology, The Ohio State University, 1835 Neil Ave, Columbus, OH 43210, USA; Group in Behavioral Neuroendocrinology, The Ohio State University, Columbus OH, USA.
| |
Collapse
|
24
|
Hübner S, Sunny DE, Pöhlke C, Ruhnau J, Vogelgesang A, Reich B, Heckmann M. Protective Effects of Fetal Zone Steroids Are Comparable to Estradiol in Hyperoxia-Induced Cell Death of Immature Glia. Endocrinology 2017; 158:1419-1435. [PMID: 28323976 DOI: 10.1210/en.2016-1763] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/24/2017] [Indexed: 12/18/2022]
Abstract
Impaired neurodevelopment in preterm infants is caused by prematurity itself; however, hypoxia/ischemia, inflammation, and hyperoxia contribute to the extent of impairment. Because preterm birth is accompanied by a dramatic decrease in 17β-estradiol (E2) and progesterone, preliminary clinical studies have been carried out to substitute these steroids in preterm infants; however, they failed to confirm significantly improved neurologic outcomes. We therefore hypothesized that the persistently high postnatal production of fetal zone steroids [mainly dehydroepiandrosterone (DHEA)] until term could interfere with E2-mediated protection. We investigated whether E2 could reduce hyperoxia-mediated apoptosis in three immature glial cell types and detected the involved receptors. Thereafter, we investigated protection by the fetal zone steroids DHEA, 16α-hydroxy-DHEA, and androstenediol. For DHEA, the involved receptors were evaluated. We examined aromatases, which convert fetal zone steroids into more estrogenic compounds. Finally, cotreatment was compared against single hormone treatment to investigate synergism. In all cell types, E2 and fetal zone steroids resulted in significant dose-dependent protection, whereas the mediating receptors differed. The neuroprotection by fetal zone steroids highly depended on the cell type-specific expression of aromatases, the receptor repertoire, and the potency of the fetal zone steroids toward these receptors. No synergism in fetal zone steroid and E2 cotreatment was detected in two of three cell types. Therefore, E2 supplementation may not be beneficial with respect to neuroprotection because fetal zone steroids circulate in persistently high concentrations until term in preterm infants. Hence, a refined experimental model for preterm infants is required to investigate potential treatments.
Collapse
Affiliation(s)
- Stephanie Hübner
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, 17457 Greifswald, Germany
| | - Donna E Sunny
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, 17457 Greifswald, Germany
| | - Christine Pöhlke
- Section of Neuroimmunology, Department of Neurology, University Medicine Greifswald, 17457 Greifswald, Germany
| | - Johanna Ruhnau
- Section of Neuroimmunology, Department of Neurology, University Medicine Greifswald, 17457 Greifswald, Germany
| | - Antje Vogelgesang
- Section of Neuroimmunology, Department of Neurology, University Medicine Greifswald, 17457 Greifswald, Germany
| | - Bettina Reich
- Pediatric Heart Center, Department of Pediatric Cardiology, Justus Liebig University, 35385 Giessen, Germany
| | - Matthias Heckmann
- Department of Neonatology and Pediatric Intensive Care, University Medicine Greifswald, 17457 Greifswald, Germany
| |
Collapse
|
25
|
Zhang H, Lu L, Zhu M, Zhang F, Sheng X, Yuan Z, Han Y, Watanabe G, Taya K, Weng Q. Seasonal expression of P450arom and estrogen receptors in scented glands of muskrats (Ondatra zibethicus). Am J Physiol Regul Integr Comp Physiol 2017; 312:R380-R387. [DOI: 10.1152/ajpregu.00458.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/13/2016] [Accepted: 12/24/2016] [Indexed: 02/08/2023]
Abstract
Male muskrats have one pair of scented glands that grow and involute annually. To investigate the annual changes in the scented gland, we measured the expressions of aromatase cytochrome P-450 (P450arom) and estrogen receptors (ERs) in the scented glands. P450arom was expressed in glandular cells and epithelial cells in the scented glands during the breeding season, and only in glandular cells during the nonbreeding season. ERα and ERβ were also detected in different types of cells in the scented gland during the breeding and nonbreeding seasons. Both mRNA and protein levels of P450arom, ERα, and ERβ were higher in the scented glandular tissues during the breeding season than those during the nonbreeding season. In addition, small RNA sequencing showed that the predicted targets of the significantly changed microRNAs might be the genes encoding P450arom and ERs. In conclusion, the seasonal changes in the expression of P450arom and ERs may be involved in the regulation of scented gland functions.
Collapse
Affiliation(s)
- Haolin Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Lu Lu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Manyu Zhu
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Fengwei Zhang
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Xia Sheng
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Zhengrong Yuan
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Yingying Han
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| | - Gen Watanabe
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Kazuyoshi Taya
- Laboratory of Veterinary Physiology, Department of Veterinary Medicine, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Qiang Weng
- Laboratory of Animal Physiology, College of Biological Sciences and Technology, Beijing Forestry University, Beijing Peoples Republic of China; and
| |
Collapse
|
26
|
Zhao Y, Yu Y, Zhang Y, He L, Qiu L, Zhao J, Liu M, Zhang J. Letrozole regulates actin cytoskeleton polymerization dynamics in a SRC-1 dependent manner in the hippocampus of mice. J Steroid Biochem Mol Biol 2017; 167:86-97. [PMID: 27866972 DOI: 10.1016/j.jsbmb.2016.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/24/2016] [Accepted: 11/16/2016] [Indexed: 12/21/2022]
Abstract
In the hippocampus, local estrogens (E2) derived from testosterone that is catalyzed by aromatase play important roles in the regulation of hippocampal neural plasticity, but the underlying mechanisms remain unclear. The actin cytoskeleton contributes greatly to hippocampal synaptic plasticity; however, whether it is regulated by local E2 and the related mechanisms remain to be elucidated. In this study, we first examined the postnatal developmental profiles of hippocampal aromatase and specific proteins responsible for actin cytoskeleton dynamics. Then we used aromatase inhibitor letrozole (LET) to block local E2 synthesis and examined the changes of these proteins and steroid receptor coactivator-1 (SRC-1), the predominant coactivator for steroid nuclear receptors. Finally, SRC-1 specific RNA interference was used to examine the effects of SRC-1 on the expression of these actin remodeling proteins. The results showed a V-type profile for aromatase and increased profiles for actin cytoskeleton proteins in both male and female hippocampus without obvious sex differences. LET treatment dramatically decreased the F-actin/G-actin ratio, the expression of Rictor, phospho-AKT (ser473), Profilin-1, phospho-Cofilin (Ser3), and SRC-1 in a dose-dependent manner. In vitro studies demonstrated that LET induced downregulation of these proteins could be reversed by E2, and E2 induced increase of these proteins were significantly suppressed by SRC-1 shRNA interference. These results for the first time clearly demonstrated that local E2 inhibition could induce aberrant actin polymerization; they also showed an important role of SRC-1 in the mediation of local E2 action on hippocampal synaptic plasticity by regulation of actin cytoskeleton dynamics.
Collapse
Affiliation(s)
- Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yanlan Yu
- Student Brigade, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Linli Qiu
- School of Nursing, Third Military Medical University, Chongqing 400038, China; Department of Nursing, Sichuan Nursing Vocational College, Chengdu 610100, China
| | - Jikai Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Mengying Liu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
27
|
Ponnusamy S, Tran QT, Harvey I, Smallwood HS, Thiyagarajan T, Banerjee S, Johnson DL, Dalton JT, Sullivan RD, Miller DD, Bridges D, Narayanan R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue. FASEB J 2016; 31:266-281. [PMID: 27733447 DOI: 10.1096/fj.201600787rr] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023]
Abstract
Most satiety-inducing obesity therapeutics, despite modest efficacy, have safety concerns that underscore the need for effective peripherally acting drugs. An attractive therapeutic approach for obesity is to optimize/maximize energy expenditure by increasing energy-utilizing thermogenic brown adipose tissue. We used in vivo and in vitro models to determine the role of estrogen receptor β (ER-β) and its ligands on adipose biology. RNA sequencing and metabolomics were used to determine the mechanism of action of ER-β and its ligands. Estrogen receptor β (ER-β) and its selective ligand reprogrammed preadipocytes and precursor stem cells into brown adipose tissue and increased mitochondrial respiration. An ER-β-selective ligand increased markers of tricarboxylic acid-dependent and -independent energy biogenesis and oxygen consumption in mice without a concomitant increase in physical activity or food consumption, all culminating in significantly reduced weight gain and adiposity. The antiobesity effects of ER-β ligand were not observed in ER-β-knockout mice. Serum metabolite profiles of adult lean and juvenile mice were comparable, while that of adult obese mice was distinct, indicating a possible impact of obesity on age-dependent metabolism. This phenotype was partially reversed by ER-β-selective ligand. These data highlight a new role for ER-β in adipose biology and its potential to be a safer alternative peripheral therapeutic target for obesity.-Ponnusamy, S., Tran, Q. T., Harvey, I., Smallwood, H. S., Thiyagarajan, T., Banerjee, S., Johnson, D. L., Dalton, J. T., Sullivan, R. D., Miller, D. D., Bridges, D., Narayanan, R. Pharmacologic activation of estrogen receptor β increases mitochondrial function, energy expenditure, and brown adipose tissue.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Quynh T Tran
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Innocence Harvey
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Thirumagal Thiyagarajan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Daniel L Johnson
- Molecular Informatics Core, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James T Dalton
- Preclinical Research and Development, GTx, Incorporated, Memphis, Tennessee, USA
| | - Ryan D Sullivan
- Department of Comparative Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; and
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Dave Bridges
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA; .,West Cancer Center, Memphis, Tennessee, USA
| |
Collapse
|
28
|
Arambula SE, Belcher SM, Planchart A, Turner SD, Patisaul HB. Impact of Low Dose Oral Exposure to Bisphenol A (BPA) on the Neonatal Rat Hypothalamic and Hippocampal Transcriptome: A CLARITY-BPA Consortium Study. Endocrinology 2016; 157:3856-3872. [PMID: 27571134 PMCID: PMC5045502 DOI: 10.1210/en.2016-1339] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/23/2016] [Indexed: 11/19/2022]
Abstract
Bisphenol A (BPA) is an endocrine disrupting, high volume production chemical found in a variety of products. Evidence of prenatal exposure has raised concerns that developmental BPA may disrupt sex-specific brain organization and, consequently, induce lasting changes on neurophysiology and behavior. We and others have shown that exposure to BPA at doses below the no-observed-adverse-effect level can disrupt the sex-specific expression of estrogen-responsive genes in the neonatal rat brain including estrogen receptors (ERs). The present studies, conducted as part of the Consortium Linking Academic and Regulatory Insights of BPA Toxicity program, expanded this work by examining the hippocampal and hypothalamic transcriptome on postnatal day 1 with the hypothesis that genes sensitive to estrogen and/or sexually dimorphic in expression would be altered by prenatal BPA exposure. NCTR Sprague-Dawley dams were gavaged from gestational day 6 until parturition with BPA (0-, 2.5-, 25-, 250-, 2500-, or 25 000-μg/kg body weight [bw]/d). Ethinyl estradiol was used as a reference estrogen (0.05- or 0.5-μg/kg bw/d). Postnatal day 1 brains were microdissected and gene expression was assessed with RNA-sequencing (0-, 2.5-, and 2500-μg/kg bw BPA groups only) and/or quantitative real-time PCR (all exposure groups). BPA-related transcriptional changes were mainly confined to the hypothalamus. Consistent with prior observations, BPA induced sex-specific effects on hypothalamic ERα and ERβ (Esr1 and Esr2) expression and hippocampal and hypothalamic oxytocin (Oxt) expression. These data demonstrate prenatal BPA exposure, even at doses below the current no-observed-adverse-effect level, can alter gene expression in the developing brain.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Biological Sciences (S.E.A., S.M.B., A.P., H.B.P.), Keck Center for Behavioral Biology (S.E.A., H.B.P.), and Center for Human Health and the Environment (S.E.A., S.M.B., A.P., H.B.P.), North Carolina State University, Raleigh, North Carolina 27695; and Department of Public Health Sciences (S.D.T.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Scott M Belcher
- Department of Biological Sciences (S.E.A., S.M.B., A.P., H.B.P.), Keck Center for Behavioral Biology (S.E.A., H.B.P.), and Center for Human Health and the Environment (S.E.A., S.M.B., A.P., H.B.P.), North Carolina State University, Raleigh, North Carolina 27695; and Department of Public Health Sciences (S.D.T.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Antonio Planchart
- Department of Biological Sciences (S.E.A., S.M.B., A.P., H.B.P.), Keck Center for Behavioral Biology (S.E.A., H.B.P.), and Center for Human Health and the Environment (S.E.A., S.M.B., A.P., H.B.P.), North Carolina State University, Raleigh, North Carolina 27695; and Department of Public Health Sciences (S.D.T.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Stephen D Turner
- Department of Biological Sciences (S.E.A., S.M.B., A.P., H.B.P.), Keck Center for Behavioral Biology (S.E.A., H.B.P.), and Center for Human Health and the Environment (S.E.A., S.M.B., A.P., H.B.P.), North Carolina State University, Raleigh, North Carolina 27695; and Department of Public Health Sciences (S.D.T.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| | - Heather B Patisaul
- Department of Biological Sciences (S.E.A., S.M.B., A.P., H.B.P.), Keck Center for Behavioral Biology (S.E.A., H.B.P.), and Center for Human Health and the Environment (S.E.A., S.M.B., A.P., H.B.P.), North Carolina State University, Raleigh, North Carolina 27695; and Department of Public Health Sciences (S.D.T.), University of Virginia School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
29
|
Green MR, McCormick CM. Sex and stress steroids in adolescence: Gonadal regulation of the hypothalamic-pituitary-adrenal axis in the rat. Gen Comp Endocrinol 2016; 234:110-6. [PMID: 26851306 DOI: 10.1016/j.ygcen.2016.02.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/02/2016] [Indexed: 01/19/2023]
Abstract
This review provides an overview of the current understanding of the role of the hypothalamic-pituitary-gonadal (HPG) axis in regulating the hypothalamic-pituitary-adrenal (HPA) axis response to stressors. HPA function is influenced by both organizational (programming) and activational effects of gonadal hormones. Typically, in adult rats, estradiol increases and androgens decrease the HPA response to stressors, thereby contributing to sex differences in HPA function, and sensitivity of the HPA axis to gonadal steroids is in part determined by exposure to these hormones in early development. Although developmental differences in HPA function are well characterized, the extent to which gonadal steroids contribute to age differences in HPA function is not well understood. Deficits in the understanding of the relationships between the HPA and HPG axes are greatest for the adolescent period of development. The critical outstanding questions are, when do gonadal hormones begin to regulate HPA function in adolescence, and what mechanisms precipitate change in sensitivity of the HPA axis to the HPG axis at this stage of life.
Collapse
Affiliation(s)
- Matthew R Green
- Department of Psychology, Brock University, 500 Glenridge Avenue, St. Catharines, Ontario L2S 3A1, Canada
| | - Cheryl M McCormick
- Department of Psychology, Brock University, 500 Glenridge Avenue, St. Catharines, Ontario L2S 3A1, Canada; Centre for Neuroscience, Brock University, 500 Glenridge Avenue, St. Catharines, Ontario L2S 3A1, Canada.
| |
Collapse
|
30
|
Tuscher JJ, Szinte JS, Starrett JR, Krentzel AA, Fortress AM, Remage-Healey L, Frick KM. Inhibition of local estrogen synthesis in the hippocampus impairs hippocampal memory consolidation in ovariectomized female mice. Horm Behav 2016; 83:60-67. [PMID: 27178577 PMCID: PMC4915975 DOI: 10.1016/j.yhbeh.2016.05.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 05/02/2016] [Indexed: 01/29/2023]
Abstract
The potent estrogen 17β-Estradiol (E2) plays a critical role in mediating hippocampal function, yet the precise mechanisms through which E2 enhances hippocampal memory remain unclear. In young adult female rodents, the beneficial effects of E2 on memory are generally attributed to ovarian-synthesized E2. However, E2 is also synthesized in the adult brain in numerous species, where it regulates synaptic plasticity and is synthesized in response to experiences such as exposure to females or conspecific song. Although de novo E2 synthesis has been demonstrated in rodent hippocampal cultures, little is known about the functional role of local E2 synthesis in mediating hippocampal memory function. Therefore, the present study examined the role of hippocampal E2 synthesis in hippocampal memory consolidation. Using bilateral dorsal hippocampal infusions of the aromatase inhibitor letrozole, we first found that blockade of dorsal hippocampal E2 synthesis impaired hippocampal memory consolidation. We next found that elevated levels of E2 in the dorsal hippocampus observed 30min after object training were blocked by dorsal hippocampal infusion of letrozole, suggesting that behavioral experience increases acute and local E2 synthesis. Finally, aromatase inhibition did not prevent exogenous E2 from enhancing hippocampal memory consolidation, indicating that hippocampal E2 synthesis is not necessary for exogenous E2 to enhance hippocampal memory. Combined, these data are consistent with the hypothesis that hippocampally-synthesized E2 is necessary for hippocampus-dependent memory consolidation in rodents.
Collapse
Affiliation(s)
- Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Julia S Szinte
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Joseph R Starrett
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Amanda A Krentzel
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ashley M Fortress
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Luke Remage-Healey
- Department of Psychological and Brain Sciences and Neuroscience and Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA, United States
| | - Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| |
Collapse
|
31
|
Paulukat L, Frintrop L, Liesbrock J, Heussen N, Johann S, Exner C, Kas MJ, Tolba R, Neulen J, Konrad K, Herpertz-Dahlmann B, Beyer C, Seitz J. Memory impairment is associated with the loss of regular oestrous cycle and plasma oestradiol levels in an activity-based anorexia animal model. World J Biol Psychiatry 2016; 17:274-84. [PMID: 27160428 DOI: 10.3109/15622975.2016.1173725] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Patients with anorexia nervosa (AN) suffer from neuropsychological deficits including memory impairments. Memory partially depends on 17β-oestradiol (E2), which is reduced in patients with AN. We assessed whether memory functions correlate with E2 plasma levels in the activity-based anorexia (ABA) rat model. METHODS Nine 4-week-old female Wistar rats were sacrificed directly after weight loss of 20-25% (acute starvation), whereas 17 animals had additional 2-week weight-holding (chronic starvation). E2 serum levels and novel object recognition tasks were tested before and after starvation and compared with 21 normally fed controls. RESULTS Starvation disrupted menstrual cycle and impaired memory function, which became statistically significant in the chronic state (oestrous cycle (P < 0.001), E2 levels (P = 0.011) and object recognition memory (P = 0.042) compared to controls). E2 reduction also correlated with the loss of memory in the chronic condition (r = 0.633, P = 0.020). CONCLUSIONS Our results demonstrate that starvation reduces the E2 levels which are associated with memory deficits in ABA rats. These effects might explain reduced memory capacity in patients with AN as a consequence of E2 deficiency and the potentially limited effectiveness of psychotherapeutic interventions in the starved state. Future studies should examine whether E2 substitution could prevent cognitive deficits and aid in earlier readiness for therapy.
Collapse
Affiliation(s)
- Lisa Paulukat
- a Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Aachen, RWTH Aachen University , Aachen , Germany ;,b Institute of Neuroanatomy , RWTH Aachen University , Aachen , Germany
| | - Linda Frintrop
- b Institute of Neuroanatomy , RWTH Aachen University , Aachen , Germany
| | - Johanna Liesbrock
- a Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Aachen, RWTH Aachen University , Aachen , Germany ;,b Institute of Neuroanatomy , RWTH Aachen University , Aachen , Germany
| | - Nicole Heussen
- c Department of Medical Statistics , University Hospital Aachen, RWTH Aachen University , Aachen , Germany
| | - Sonja Johann
- b Institute of Neuroanatomy , RWTH Aachen University , Aachen , Germany
| | - Cornelia Exner
- d Department of Animal Physiology , Philipps-University Marburg , Marburg , Germany
| | - Martien J Kas
- e Department of Translational Neuroscience, Brain Center Rudolf Magnus , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Rene Tolba
- f Institute for Laboratory Animal Science and Experimental Surgery , University Hospital Aachen, RWTH Aachen University , Aachen , Germany
| | - Joseph Neulen
- g Department of Gynecological Endocrinology and Reproductive Medicine , University Hospital, RWTH Aachen University , Aachen , Germany
| | - Kerstin Konrad
- a Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Aachen, RWTH Aachen University , Aachen , Germany
| | - Beate Herpertz-Dahlmann
- a Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Aachen, RWTH Aachen University , Aachen , Germany
| | - Cordian Beyer
- b Institute of Neuroanatomy , RWTH Aachen University , Aachen , Germany
| | - Jochen Seitz
- a Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy , University Hospital Aachen, RWTH Aachen University , Aachen , Germany
| |
Collapse
|
32
|
Chang H, Wang M, Xia W, Chen T, Huo W, Mao Z, Zhu Y, Li Y, Xu S. Perinatal exposure to low-dose bisphenol A disrupts learning/memory and DNA methylation of estrogen receptor alpha in the hippocampus. Toxicol Res (Camb) 2016; 5:828-835. [PMID: 30090393 PMCID: PMC6060734 DOI: 10.1039/c5tx00449g] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 01/31/2016] [Indexed: 11/21/2022] Open
Abstract
Developmental exposure to bisphenol A (BPA) has been indicated to pose long-lasting effects on brain development and behaviors in adulthood. Previous studies have also shown that BPA may disrupt the epigenetic programming of genes in the brain. Here, we focused on investigating the effects of perinatal exposure to low-dose BPA on learning/memory function and emotional regulation, as well as the associated molecular events. Pregnant Sprague-Dawley (SD) rats were treated with control corn oil or BPA (40 μg kg-1 per day) throughout gestation and lactation. Morris water maze (MWM) and elevated plus maze (EPM) were used to evaluate learning/memory and anxiety-like behaviors at postnatal day (PND) 60 and 85 respectively. The expression level of mRNA for estrogen receptors (ER), ERα and ERβ, in the hippocampus and the serum corticosterone level were determined, as well as the DNA methylation status of the ERα gene promoter. Perinatal exposure to BPA prolonged the escape latency independent of gender, and decreased the percentage of time spent in the target quadrant when examined in the MWM task. While no substantial alteration was observed in the EPM test, the serum corticosterone level was altered in a gender-specific manner. BPA also decreased the expression of mRNA for ERα in the hippocampus, along with elevated DNA methylation of the ERα gene promoter. These results suggest that perinatal exposure to BPA impairs learning/memory function and elevated DNA methylation of the ERα gene in the hippocampus may be involved.
Collapse
Affiliation(s)
- Huailong Chang
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Mu Wang
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Wei Xia
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Tian Chen
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Wenqian Huo
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Zhenxing Mao
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Yingshuang Zhu
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Yuanyuan Li
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| | - Shunqing Xu
- Key Laboratory of Environment and Health , Ministry of Education & Ministry of Environmental Protection , and State Key Laboratory of Environmental Health , School of Public Health , Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430030 , China . ; ; ; Tel: +86-27-83657705, +86-27-83693417
| |
Collapse
|
33
|
Qiu L, Zhao Y, Guo Q, Zhang Y, He L, Li W, Zhang J. Dose-dependent regulation of steroid receptor coactivator-1 and steroid receptors by testosterone propionate in the hippocampus of adult male mice. J Steroid Biochem Mol Biol 2016; 156:23-31. [PMID: 26607693 DOI: 10.1016/j.jsbmb.2015.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 01/08/2023]
Abstract
Androgens have been proposed to play important roles in the regulation of hippocampus function either directly, through the androgen receptor (AR), or indirectly, through estrogen receptors (ERs), after aromatization into estradiol. Steroid receptor coactivator-1 (SRC-1) is present in the hippocampus of several species, and its expression is regulated by development and aging, as well as by orchidectomy and aromatase inhibitor letrozole administration, while ovariectomy only transiently downregulated hippocampal SRC-1. However, whether the expression of hippocampal SRC-1 can be directly regulated by testosterone, the principal male sex hormone, remains unclear. In the present study, we investigated the expression of hippocampal SRC-1 after orchidectomy and testosterone treatment using immunohistochemistry and Western blot analysis. We found that while hippocampal SRC-1 was significantly downregulated by orchidectomy (ORX), its expression was rescued by treatment with testosterone in a dose-dependent manner. Furthermore, we noticed that the decreased expression of hippocampal AR, ERs and the synaptic proteins GluR-1 and PSD-95 induced by ORX was also rescued by testosterone treatment in a dose-dependent manner. However, we found that hippocampal membrane estrogen receptor GPR30 and dendritic spine marker spinophilin were not altered by ORX or testosterone treatment. Together, the above results provided the first direct evidence for the androgenic regulation on hippocampal SRC-1, indicating that SRC-1 may be a direct target of androgenic regulation on the hippocampus. Furthermore, because AR and ERs can be differentially regulated by testosterone, and the transcriptional activity requires the involvement of local SRC-1, and considering the complicated regulatory pathway of each individual receptor, the converged hub regulator SRC-1 of these nuclear receptor networks is worthy of further investigation.
Collapse
Affiliation(s)
- Linli Qiu
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China; Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Yangang Zhao
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Qiang Guo
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Yuanyuan Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | - Li He
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China; Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China
| | - Wei Li
- Department of Filed Nursing, School of Nursing, Third Military Medical University, Chongqing 400038, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
Vahaba DM, Remage-Healey L. Brain estrogen production and the encoding of recent experience. Curr Opin Behav Sci 2015; 6:148-153. [PMID: 27453921 DOI: 10.1016/j.cobeha.2015.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The vertebrate central nervous system integrates cognition and behavior, and it also acts as both a source and target for steroid hormones like estrogens. Recent exploration of brain estrogen production in the context of learning and memory has revealed several common themes. First, across vertebrates, the enzyme that synthesizes estrogens is expressed in brain regions that are characterized by elevated neural plasticity and is also integral to the acquisition, consolidation, and retrieval of recent experiences. Second, measurement and manipulation of estrogens reveal that the period following recent sensory experience is linked to estrogenic signaling in brain circuits underlying both spatial and vocal learning. Local brain estrogen production within cognitive circuits may therefore be important for the acquisition and/or consolidation of memories, and new directions testing these ideas will be discussed.
Collapse
Affiliation(s)
- Daniel M Vahaba
- Neuroscience & Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA, USA 01003
| | - Luke Remage-Healey
- Neuroscience & Behavior Program, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA, USA 01003
| |
Collapse
|
35
|
Cisternas CD, Tome K, Caeiro XE, Dadam FM, Garcia-Segura LM, Cambiasso MJ. Sex chromosome complement determines sex differences in aromatase expression and regulation in the stria terminalis and anterior amygdala of the developing mouse brain. Mol Cell Endocrinol 2015; 414:99-110. [PMID: 26231585 DOI: 10.1016/j.mce.2015.07.027] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/24/2015] [Accepted: 07/25/2015] [Indexed: 10/23/2022]
Abstract
Aromatase, which converts testosterone in estradiol, is involved in the generation of brain sex dimorphisms. Here we used the "four core genotypes" mouse model, in which the effect of gonadal sex and sex chromosome complement is dissociated, to determine if sex chromosomes influence the expression of brain aromatase. The brain of 16 days old XY mouse embryos showed higher aromatase expression in the stria terminalis and the anterior amygdaloid area than the brain of XX embryos, independent of gonadal sex. Furthermore, estradiol or dihydrotestosterone increased aromatase expression in cultures of anterior amygdala neurons derived from XX embryos, but not in those derived from XY embryos. This effect was also independent of gonadal sex. The expression of other steroidogenic molecules, estrogen receptor-α and androgen receptor was not influenced by sex chromosomes. In conclusion, sex chromosomes determine sex dimorphisms in aromatase expression and regulation in the developing mouse brain.
Collapse
Affiliation(s)
- Carla D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Karina Tome
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ximena E Caeiro
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia M Dadam
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - María J Cambiasso
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina; Departamento de Biología Bucal, Facultad de Odontología - Universidad Nacional de Córdoba, Córdoba, Argentina.
| |
Collapse
|
36
|
Mogi K, Takanashi H, Nagasawa M, Kikusui T. Sex differences in spatiotemporal expression of AR, ERα, and ERβ mRNA in the perinatal mouse brain. Neurosci Lett 2014; 584:88-92. [PMID: 25459284 DOI: 10.1016/j.neulet.2014.10.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/24/2014] [Accepted: 10/14/2014] [Indexed: 11/20/2022]
Abstract
It has been shown that every masculinized function might be organized by a particular contribution of androgens vs. estrogens in a critical time window. Here, we aimed to investigate the sex differences in brain testosterone levels and in the spatiotemporal dynamics of steroid receptor mRNA expression in perinatal mice, by using enzyme immunoassay and real-time PCR, respectively. We found that testosterone levels in the forebrain transiently increased around birth in male mice. During the perinatal period, levels of androgen receptor mRNA in the hypothalamus (hypo) and prefrontal cortex (PFC) were higher in male mice than in female mice. Estrogen receptor α (ERα) mRNA levels in the hypo and hippocampus were higher in male mice than in female mice before birth. In contrast, ERβ mRNA expression in the PFC was higher in female mice immediately after birth. These spatiotemporal sex differences in steroid receptor expression might contribute to organizing sex differences of not only reproductive function, but also anxiety, stress responses, and cognition in mice.
Collapse
Affiliation(s)
- Kazutaka Mogi
- Companion Animal Research, School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| | - Haruka Takanashi
- Companion Animal Research, School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| | - Miho Nagasawa
- Companion Animal Research, School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan
| | - Takefumi Kikusui
- Companion Animal Research, School of Veterinary Medicine, Azabu University, Sagamihara 252-5201, Japan.
| |
Collapse
|
37
|
Xu XB, He Y, Song C, Ke X, Fan SJ, Peng WJ, Tan R, Kawata M, Matsuda KI, Pan BX, Kato N. Bisphenol a regulates the estrogen receptor alpha signaling in developing hippocampus of male rats through estrogen receptor. Hippocampus 2014; 24:1570-80. [DOI: 10.1002/hipo.22336] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2014] [Indexed: 01/22/2023]
Affiliation(s)
- Xiao-Bin Xu
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
| | - Ye He
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
- Department of Pharmacology; Nanchang University; Nanchang 330031 China
| | - Chen Song
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
| | - Xin Ke
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
| | - Shi-Jun Fan
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
| | - Wei-Jie Peng
- Department of Pharmacology; Nanchang University; Nanchang 330031 China
| | - Ruei Tan
- Department of Psychiatry; School of Medicine; Showa University. 6-11-11; Tokyo 157-8577 Japan
| | - Mitsuhiro Kawata
- Department of Anatomy and Neurobiology; Kyoto Prefectural University of Medicine; Kyoto 602-8566 Japan
| | - Ken-Ichi Matsuda
- Department of Anatomy and Neurobiology; Kyoto Prefectural University of Medicine; Kyoto 602-8566 Japan
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorder; Institute of Life Science, Nanchang University; Nanchang 330031 China
| | - Nobumasa Kato
- Department of Psychiatry; School of Medicine; Showa University. 6-11-11; Tokyo 157-8577 Japan
| |
Collapse
|
38
|
Identification of sexually dimorphic genes in the neonatal mouse cortex and hippocampus. Brain Res 2014; 1562:23-38. [PMID: 24661915 DOI: 10.1016/j.brainres.2014.03.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 03/08/2014] [Accepted: 03/15/2014] [Indexed: 01/01/2023]
Abstract
The cerebral cortex and hippocampus are important for the control of cognitive functions and social behaviors, many of which are sexually dimorphic and tightly regulated by gonadal steroid hormones via activation of their respective nuclear receptors. As different levels of sex steroid hormones are present between the sexes during early development and their receptors act as transcription factors to regulate gene expression, we hypothesize that sexually dimorphic gene expression in the developing mouse cortex and hippocampus might result in sex differences in brain structures and neural circuits governing distinct behaviors between the sexes as adults. To test our hypothesis, we used gene expression microarrays to identify 90 candidate genes differentially expressed in the neonatal cortex/hippocampus between male and female mice, including 55 male-biased and 35 female-biased genes. Among these genes, sexually dimorphic expression of eight sex chromosome genes was confirmed by reverse transcription with quantitative PCR (RT-qPCR), including three located on the X chromosome (Xist, Eif2s3x, and Kdm6a), three on the Y chromosome (Ddx3y, Eif2s3y, and Kdm5d), and two in the pseudoautosomal region of the X and Y chromosomes (Erdr1 and Mid1). In addition, five autosomal genes (Cd151, Dab2, Klk8, Meg3, and Prkdc) were also validated for their sexually dimorphic expression in the neonatal mouse cortex/hippocampus. Gene Ontology annotation analysis suggests that many of these sexually dimorphic genes are involved in histone modifications, cell proliferation/death, androgen/estrogen signaling pathways, and synaptic organization, and these biological processes have been implicated in differential neural development, cognitive function, and neurological diseases between the sexes.
Collapse
|
39
|
Bidirectional modulatory effect of 17β-estradiol on NMDA receptors via ERα and ERβ in the dentate gyrus of juvenile male rats. Neuropharmacology 2013; 75:262-73. [PMID: 23954493 DOI: 10.1016/j.neuropharm.2013.07.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/13/2013] [Accepted: 07/23/2013] [Indexed: 11/22/2022]
Abstract
The neurosteroid 17β-estradiol (E2) is synthesized by aromatase in both male and female hippocampi and is known to modulate hippocampal synaptic functions. However, as some contradictory findings regarding the modulatory effects of E2 have been reported in the literature, its physiological role and mechanism of action in the hippocampus remain controversial. Our recent study showed that a low E2 dose (1 nM) increased the amplitude of NMDA receptor-mediated EPSCs (NMDAR-EPSCs) and lowered the threshold for the induction of NMDA receptor-dependent long-term potentiation (NMDAR-LTP), while a high E2 dose (7 nM) exerted opposite effects in the dentate gyrus of juvenile male rat hippocampal slices. The present study is a follow-up that explores the underlying mechanism of this bidirectional effect of E2. We found that the ERα agonist PPT reproduced the actions of the low E2 dose on NMDAR-EPSCs and NMDAR-LTP, while the ERβ agonist DPN reproduced the actions of the high E2 dose. Moreover, PPT, but not DPN, restored the decrease in NMDAR-EPSCs induced by the aromatase inhibitor letrozole, suggesting that E2 synthesized constitutively in the hippocampus enhances NMDA receptor function via ERα. The PPT-induced enhancement in NMDAR-EPSCs was mediated by Src family kinase, but was not caused by NR2B modulation. These findings demonstrate that E2 exerts condition-dependent bidirectional effects on NMDA receptor-mediated transmission and, thus, synaptic plasticity via ERα and ERβ in the dentate gyrus of juvenile male rats.
Collapse
|
40
|
Mott NN, Pak TR. Estrogen signaling and the aging brain: context-dependent considerations for postmenopausal hormone therapy. ISRN ENDOCRINOLOGY 2013; 2013:814690. [PMID: 23936665 PMCID: PMC3725729 DOI: 10.1155/2013/814690] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/21/2013] [Indexed: 02/08/2023]
Abstract
Recent clinical studies have spurred rigorous debate about the benefits of hormone therapy (HT) for postmenopausal women. Controversy first emerged based on a sharp increase in the risk of cardiovascular disease in participants of the Women's Health Initiative (WHI) studies, suggesting that decades of empirical research in animal models was not necessarily applicable to humans. However, a reexamination of the data from the WHI studies suggests that the timing of HT might be a critical factor and that advanced age and/or length of estrogen deprivation might alter the body's ability to respond to estrogens. Dichotomous estrogenic effects are mediated primarily by the actions of two high-affinity estrogen receptors alpha and beta (ER α & ER β ). The expression of the ERs can be overlapping or distinct, dependent upon brain region, sex, age, and exposure to hormone, and, during the time of menopause, there may be changes in receptor expression profiles, post-translational modifications, and protein:protein interactions that could lead to a completely different environment for E2 to exert its effects. In this review, factors affecting estrogen-signaling processes will be discussed with particular attention paid to the expression and transcriptional actions of ER β in brain regions that regulate cognition and affect.
Collapse
Affiliation(s)
- Natasha N. Mott
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| | - Toni R. Pak
- Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, 2160 S First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
41
|
Waddell J, Bowers JM, Edwards NS, Jordan CL, McCarthy MM. Dysregulation of neonatal hippocampal cell genesis in the androgen insensitive Tfm rat. Horm Behav 2013; 64:144-52. [PMID: 23747829 PMCID: PMC3753588 DOI: 10.1016/j.yhbeh.2013.05.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 05/23/2013] [Accepted: 05/29/2013] [Indexed: 12/20/2022]
Abstract
The first two weeks of life are a critical period for hippocampal development. At this time gonadal steroid exposure organizes sex differences in hippocampal sensitivity to activational effects of steroids, hippocampal cell morphology and hippocampus dependent behaviors. Our laboratory has characterized a robust sex difference in neonatal neurogenesis in the hippocampus that is mediated by estradiol. Here, we extend our knowledge of this sex difference by comparing the male and female hippocampus to the androgen insensitive testicular feminized mutant (Tfm) rat. In the neonatal Tfm rat hippocampus, fewer newly generated cells survive compared to males or females. This deficit in cell genesis is partially recovered with the potent androgen DHT, but is more completely recovered following estradiol administration. Tfm rats do not differ from males or females in the level of endogenous estradiol in the neonatal hippocampus, suggesting other mechanisms mediate a differential sensitivity to estradiol in male, female and Tfm hippocampus. We also demonstrate disrupted performance on a hippocampal-dependent contextual fear discrimination task. Tfm rats generalize fear across contexts, and do not exhibit significant loss of fear during extinction exposure. These results extend prior reports of exaggerated response to stress in Tfm rats, and following gonadectomy in normal male rats.
Collapse
Affiliation(s)
- Jaylyn Waddell
- Department of Pediatrics, University of Maryland, Baltimore, School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | |
Collapse
|
42
|
Ooishi Y, Kawato S, Hojo Y, Hatanaka Y, Higo S, Murakami G, Komatsuzaki Y, Ogiue-Ikeda M, Kimoto T, Mukai H. Modulation of synaptic plasticity in the hippocampus by hippocampus-derived estrogen and androgen. J Steroid Biochem Mol Biol 2012; 131:37-51. [PMID: 22075082 DOI: 10.1016/j.jsbmb.2011.10.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Revised: 09/27/2011] [Accepted: 10/12/2011] [Indexed: 12/29/2022]
Abstract
The hippocampus synthesizes estrogen and androgen in addition to the circulating sex steroids. Synaptic modulation by hippocampus-derived estrogen or androgen is essential to maintain healthy memory processes. Rapid actions (1-2h) of 17β-estradiol (17β-E2) occur via synapse-localized receptors (ERα or ERβ), while slow genomic E2 actions (6-48h) occur via classical nuclear receptors (ERα or ERβ). The long-term potentiation (LTP), induced by strong tetanus or theta-burst stimulation, is not further enhanced by E2 perfusion in adult rats. Interestingly, E2 perfusion can rescue corticosterone (stress hormone)-induced suppression of LTP. The long-term depression is modulated rapidly by E2 perfusion. Elevation of the E2 concentration changes rapidly the density and head structure of spines in neurons. ERα, but not ERβ, drives this enhancement of spinogenesis. Kinase networks are involved downstream of ERα. Testosterone (T) or dihydrotestosterone (DHT) also rapidly modulates spinogenesis. Newly developed Spiso-3D mathematical analysis is used to distinguish these complex effects by sex steroids and kinases. It has been doubted that the level of hippocampus-derived estrogen and androgen may not be high enough to modulate synaptic plasticity. Determination of the accurate concentration of E2, T or DHT in the hippocampus is enabled by mass-spectrometric analysis in combination with new steroid-derivatization methods. The E2 level in the hippocampus is approximately 8nM for the male and 0.5-2nM for the female, which is much higher than that in circulation. The level of T and DHT is also higher than that in circulation. Taken together, hippocampus-derived E2, T, and DHT play a major role in modulation of synaptic plasticity.
Collapse
Affiliation(s)
- Yuuki Ooishi
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bian C, Zhu K, Yang L, Lin S, Li S, Su B, Zhang J. Gonadectomy differentially regulates steroid receptor coactivator-1 and synaptic proteins in the hippocampus of adult female and male C57BL/6 mice. Synapse 2012; 66:849-57. [PMID: 22623226 DOI: 10.1002/syn.21574] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/14/2012] [Indexed: 02/06/2023]
Abstract
Hippocampus is one of the most important structures that mediates learning and memory, cognition, and mental behaviors and profoundly regulated by sex hormones in a sex-specific manner, but the mechanism of underlying sex differences regulation is still unclear. We have previously reported that in the male and female mice, steroid receptor coactivator-1 (SRC-1) and some key synaptic proteins share similar developmental profile in the hippocampus, but how circulating sex hormones affect hippocampal SRC-1 as well as these synaptic proteins remain unclear. In this study, we examined how gonad sex hormones regulate hippocampal SRC-1, synaptophysin, PSD-95, and AMPA receptor subtype GluR1 by using immunohistochemistry and Western blot. The results showed that in the female mice, ovariectomy affected hippocampal SRC-1 and GluR1 were only detected at 2 weeks post operation, then it recovered to sham level; synaptophysin was unaffected at any timepoint examined; significant decrease of PSD-95 was only detected at 4 weeks post operation. However, in the male hippocampus, SRC-1 and PSD-95 were decreased from one week and lasted to 4 weeks after orchidectomy, GluR1 decreased from 2 weeks after orchidectomy, but synaptophysin remained unchanged as in the females. Correlation analysis showed the profiles of SRC-1 were positively correlated with GluR1 of the females, PSD-95 and GluR1 of the males, respectively. The above results suggested a distinct regulatory mode between female and male gonad hormones in the regulation of hippocampal SRC-1 and synaptic proteins, which may be one of the mechanisms contributing to the dimorphism of hippocampus during development and ageing.
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Bian C, Zhu K, Guo Q, Xiong Y, Cai W, Zhang J. Sex differences and synchronous development of steroid receptor coactivator-1 and synaptic proteins in the hippocampus of postnatal female and male C57BL/6 mice. Steroids 2012; 77:149-56. [PMID: 22085911 DOI: 10.1016/j.steroids.2011.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 10/25/2011] [Accepted: 11/02/2011] [Indexed: 12/13/2022]
Abstract
The structure and function including synaptic plasticity of the hippocampus are deeply affected by steroids in a sex-dependant manner, these processes are believed to be mediated by steroid receptors though their coactivators. Our previous studies have reported the developmental profiles of steroid receptor coactivator-1 (SRC-1) and PSD-95 in the hippocampus of postnatal female rats and the sex-differences of SRC-1 immunoreactivities in the brain of adult mice. However, whether there are any sex differences about postnatal development of SRC-1 and synaptic proteins in the hippocampus remain unclear. In this study, we investigated the postnatal profile of SRC-1 and key synaptic protein synaptophysin (SYN), PSD-95 and GluR1 in the hippocampus of female and male mice using immunohistochemistry and Western blot. The results showed that in the female hippocampus, the highest levels of SRC-1 were detected at P14, SYN and GluR1 at P30 and PSD-95 at P60; while in the males, the highest levels of SRC-1, SYN and GluR1 were detected at P30, and PSD-95 at P60. Female hippocampus tended to have higher levels of SRC-1, SYN and GluR1 before P30 and PSD-95 before P14; while male hippocampus have higher levels of PSD-95 at P14, P60 and GluR1 at P0. Correlation analysis showed the profiles of SRC-1 were highly correlated with each synaptic protein. The above results showed that in the hippocampus, except some minor sex differences detected at some time-point examined, females and males shared similar postnatal developmental profile and SRC-1 may be deeply involved in the regulation of hippocampal synaptogenesis.
Collapse
Affiliation(s)
- Chen Bian
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
45
|
Mendoza-Garcés L, Mendoza-Rodríguez CA, Jiménez-Trejo F, Picazo O, Rodríguez MC, Cerbón M. Differential expression of estrogen receptors in two hippocampal regions during the estrous cycle of the rat. Anat Rec (Hoboken) 2011; 294:1913-9. [PMID: 21972199 DOI: 10.1002/ar.21247] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/28/2010] [Indexed: 12/29/2022]
Abstract
In the hippocampus, estrogens increase dendritic arborization, long-term potentiation, neuroprotection, and participate in many functions related with learning, memory, and affective behaviors. The presence of both estrogen receptors alpha (ERα) and beta (ERβ) isoforms has been described in the hippocampus where they play different physiological roles. The aim of this study was to investigate, by using both techniques immunohistochemistry and Western Blot, the expression pattern of ERα and ERβ in the hippocampus of the rat along the estrous cycle. Western blot analysis was used to confirm the specificity of the antibodies used against ERα and ERβ and its relative content in the hippocampus. Results from immunohistochemical studies indicate that ERβ expression increased more than the ERα in CA1 and CA3 regions during all phases of the estrous cycle. ERβ immunoreactivity was mainly located in the nucleus and predominantly expressed in CA1 during estrous and metestrus, and in CA3 during diestrus. ERα was more abundant during estrous in comparison to other phases of the cycle in CA1 region, while it was more abundant during metestrus in CA3. Interestingly, the immunolocalization of ERα subtype was both cytoplasmic and nuclear. The overall results indicate that there is a differential expression, cellular localization, and distribution of both ER subtypes in CA1 and CA3 regions, suggesting different roles for these two receptors in the hippocampus along the estrous cycle.
Collapse
Affiliation(s)
- Luciano Mendoza-Garcés
- Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, México, Distrito Federal, México
| | | | | | | | | | | |
Collapse
|
46
|
Lu L, Zhang H, Lv N, Ma X, Tian L, Hu X, Liu S, Xu M, Weng Q, Watanabe G, Taya K. Immunolocalization of Androgen Receptor, Aromatase Cytochrome P450, Estrogen Receptor Alpha and Estrogen Receptor Beta Proteins during the Breeding Season in Scent Glands of Muskrats (Ondatra zibethicus). Zoolog Sci 2011; 28:727-32. [DOI: 10.2108/zsj.28.727] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
47
|
O'Grady SP, Caprau D, Ke XR, Contreras Y, Haley S, Ermini F, Penn A, Moyer-Mileur L, McKnight R, Lane R. Intrauterine growth restriction alters hippocampal expression and chromatin structure of Cyp19a1 variants. Syst Biol Reprod Med 2010; 56:292-302. [PMID: 20662593 DOI: 10.3109/19396368.2010.490871] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We evaluated the impact of uteroplacental insufficiency (UPI), and subsequent intrauterine growth restriction (IUGR), on serum testosterone and hippocampal expression of Cyp19a1 variants and aromatase in rats. Additionally, we determined UPI induced histone modification of the promoter regions of Cyp19a1 variants using chromatin immunoprecipitation. Cyp19a1 is the gene encoding the protein aromatase, that catalyzes the biosynthesis of estrogens from androgens and is necessary for masculinization of the brain. IUGR was induced via bilateral uterine artery. UPI increased serum testosterone in day of life 0 (D(0)) and day of life 21 (D(21)) IUGR males to 224% and 299% of control values, respectively. While there was no significant impact of UPI on testosterone in D(0) females, testosterone in D(21) IUGR females was 187% of controls. Cyp19a1 variant 1.f and variant II are expressed in the rat hippocampus at D(0) and D(21). UPI significantly reduced expression of Cyp19a1 variant 1.f in D(0) males, with no impact in females. Similarly at D(0), UPI reduced expression of aromatase, the protein encoded by Cyp19a1, in males. Dimethylation of H3K4 was increased in the promoter region of variant 1.f (P1.f) and trimethylation of H3K4 was decreased in the promoter region of variant II (PII). At D(21), dimethylation of H3K4 is significantly reduced in PII of IUGR males. We conclude that UPI increases serum testosterone and reduces Cyp19a1 variant 1.f expression in the hippocampus of D(0) IUGR males. Additionally, UPI alters the chromatin structure of CYP19a1 at both D(0) and D(21).
Collapse
|
48
|
Kimoto T, Ishii H, Higo S, Hojo Y, Kawato S. Semicomprehensive analysis of the postnatal age-related changes in the mRNA expression of sex steroidogenic enzymes and sex steroid receptors in the male rat hippocampus. Endocrinology 2010; 151:5795-806. [PMID: 21047951 DOI: 10.1210/en.2010-0581] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Although sex steroids play a crucial role in the postnatal brain development, the age-related changes in the hippocampal steroidogenesis remain largely unknown. We performed comprehensive investigations for the mRNA expressions of 26 sex steroidogenic enzymes/proteins and three sex steroid receptors in the male rat hippocampus, at the ages of postnatal day (PD) 1, PD4, PD7, PD10, PD14, 4 wk, and 12 wk (adult), by RT-PCR/Southern blotting analysis. The relative expression levels of these enzymes/receptors at PD1 were Srd5a1 > Star > Ar ∼ Hsd17b4 ∼ Hsd17b1 ∼ Hsd17b7 ∼ Esr1 ∼ Srd5a2 > Hsd17b3 > Esr2 > Cyp11a1 > Cyp17a1 > Cyp19a1 ∼ Hsd17b2 > 3β-hydroxysteroid dehydrogenase I. The mRNA levels of essential enzymes for progesterone/testosterone/estradiol metabolisms (Cyp17a1, Hsd17b7, and Cyp19a1) were approximately constant between PD1 and PD14 and then declined toward the adult levels. Cyp11a1 increased during PD4-PD14 and then considerably decreased toward the adult level (∼8% of PD1). Hsd17b1, Hsd17b2, and 3β-hydroxysteroid dehydrogenase I mRNA decreased approximately monotonously. Hsd17b3 increased to approximately 200% of PD1 during PD4-PD14 and was maintained at this high level. The 5α-reductase mRNA was maintained constant (Srd5a1) or decreased monotonically (Srd5a2) toward the adult level. The Esr1 level peaked at PD4 and decreased toward the adult level, whereas Ar greatly increased during PD1-PD14 and was maintained at this high level. The Star and Hsd17b4 levels were maintained constant from neonate to adult. These results suggest that the hippocampal sex steroidogenic properties are substantially altered during the postnatal development processes, which might contribute to brain sexual maturation.
Collapse
Affiliation(s)
- Tetsuya Kimoto
- Department of Biophysics and Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo 153-8902, Japan
| | | | | | | | | |
Collapse
|
49
|
Bowers JM, Waddell J, McCarthy MM. A developmental sex difference in hippocampal neurogenesis is mediated by endogenous oestradiol. Biol Sex Differ 2010; 1:8. [PMID: 21208470 PMCID: PMC3016241 DOI: 10.1186/2042-6410-1-8] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Accepted: 11/22/2010] [Indexed: 01/09/2023] Open
Abstract
Background Oestradiol is a steroid hormone that exerts extensive influence on brain development and is a powerful modulator of hippocampal structure and function. The hippocampus is a critical brain region regulating complex cognitive and emotional responses and is implicated in the aetiology of several mental health disorders, many of which exhibit some degree of sex difference. Many sex differences in the adult rat brain are determined by oestradiol action during a sensitive period of development. We had previously reported a sex difference in rates of cell genesis in the developing hippocampus of the laboratory rat. Males generate more new cells on average than females. The current study explored the effects of both exogenous and endogenous oestradiol on this sex difference. Methods New born male and female rat pups were injected with the mitotic marker 5-bromo-2-deoxyuridine (BrdU) and oestradiol or agents that antagonize oestradiol action. The effects on cell number, proliferation, differentiation and survival were assessed at several time points. Significant differences between groups were determined by two- or thee-Way ANOVA. Results Newborn males had higher rates of cell proliferation than females. Oestradiol treatment increased cell proliferation in neonatal females, but not males, and in the CA1 region many of these cells differentiated into neurons. The increased rate of proliferation induced by neonatal oestradiol persisted until at least 3 weeks of age, suggesting an organizational effect. Administering the aromatase inhibitor, formestane, or the oestrogen receptor antagonist, tamoxifen, significantly decreased the number of new cells in males but not females. Conclusion Endogenous oestradiol increased the rate of cell proliferation observed in newborn males compared to females. This sex difference in neonatal neurogenesis may have implications for adult differences in learning strategy, stress responsivity or vulnerability to damage or disease.
Collapse
Affiliation(s)
- J Michael Bowers
- Department of Physiology, University of Maryland, Baltimore School of Medicine Baltimore, MD 21201, USA.
| | | | | |
Collapse
|
50
|
Mukai H, Kimoto T, Hojo Y, Kawato S, Murakami G, Higo S, Hatanaka Y, Ogiue-Ikeda M. Modulation of synaptic plasticity by brain estrogen in the hippocampus. Biochim Biophys Acta Gen Subj 2010; 1800:1030-44. [DOI: 10.1016/j.bbagen.2009.11.002] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Revised: 10/15/2009] [Accepted: 11/02/2009] [Indexed: 12/31/2022]
|