1
|
Nisha, Paramanik V. Neuroprotective Roles of Daidzein Through Extracellular Signal-Regulated Kinases Dependent Pathway In Chronic Unpredictable Mild Stress Mouse Model. Mol Neurobiol 2024:10.1007/s12035-024-04567-w. [PMID: 39495229 DOI: 10.1007/s12035-024-04567-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Depression is a stress-related neuropsychiatric disorder causing behavioural, biochemical, molecular dysfunctions and cognitive impairments. Previous studies suggested connection between neuropsychiatric diseases like depression with estrogen and estrogen receptors (ER). Daidzein is a phytoestrogen that functions as mammalian estrogen and regulates gene expressions through extracellular signal-regulated kinases (ERKs) dependent pathway by activating ERβ. ERβ modulates stress responses, physiological processes by activating protein kinases and plays a significant role in various neurological diseases like depression. However, significant roles of daidzein in depression involving ERK1/2, pERK1/2, and mTOR still unknown. Herein, we examined neuroprotective role of daidzein in chronic unpredictable mild stress (CUMS) mouse model. CUMS model was prepared, and placed in six groups namely, control, CUMS, CUMS vehicle, CUMS DZ (Daidzein 1 mg/kgbw, orally), CUMS PHTPP (ERβ blocker, 0.3 mg/kgbw, i..p.) and CUMS Untreated. Supplementation of daidzein to CUMS mice exhibits decrease depressive and anxiety-like behaviour, improved motor coordination and memory. Further, immunofluorescence results showed daidzein improved ERK1/2, pERK1/2 and mTOR expressions in the cortex, hippocampus and medulla of stressed mice. SOD, catalase and acetylcholinesterase levels were also improved. Blocking of ERβ with PHTPP stressed mice showed deficits in behaviour, low expression of ERK1/2, pERK1/2 and mTOR, and no significant changes in SOD, catalase and acetylcholinesterase level. Collectively, this study suggests that daidzein may ameliorate depressive and anxiety-like behaviour through ERK downregulating pathway by activating ERβ through ERK1/2, pERK1/2 and mTOR. Such study may be useful to understand daidzein dependent neuroprotection through ERβ in depression.
Collapse
Affiliation(s)
- Nisha
- Cellular and Molecular Neurobiology and Drug Targeting Laboratory Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, 484887, MP, India
| | - Vijay Paramanik
- Cellular and Molecular Neurobiology and Drug Targeting Laboratory Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, 484887, MP, India.
| |
Collapse
|
2
|
Rymbai E, Sugumar D, Chakkittukandiyil A, Kothandan R, Selvaraj D. Molecular insights into the potential effects of selective estrogen receptor β agonists in Alzheimer's and Parkinson's diseases. Cell Biochem Funct 2024; 42:e4014. [PMID: 38616346 DOI: 10.1002/cbf.4014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative disorders. Pathologically, AD and PD are characterized by the accumulation of misfolded proteins. Hence, they are also called as proteinopathy diseases. Gender is considered as one of the risk factors in both diseases. Estrogens are widely accepted to be neuroprotective in several neurodegenerative disorders. Estrogens can be produced in the central nervous system, where they are called as neurosteroids. Estrogens mediate their neuroprotective action mainly through their actions on estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ). However, ERα is mainly involved in the growth and development of the primary and secondary sexual organs in females. Hence, the activation of ERα is associated with undesired side effects such as gynecomastia and increase in the risk of breast cancer, thromboembolism, and feminization. Therefore, selective activation of ERβ is often considered to be safer. In this review, we explore the role of ERβ in regulating the expression and functions of AD- and PD-associated genes. Additionally, we discuss the association of these genes with the amyloid-beta peptide (Aβ) and α-synuclein mediated toxicity. Ultimately, we established a correlation between the importance of ERβ activation and the process underlying ERβ's neuroprotective mechanisms in AD and PD.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Amritha Chakkittukandiyil
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Ram Kothandan
- Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, India
| | - Divakar Selvaraj
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| |
Collapse
|
3
|
Gorman-Sandler E, Wood G, Cloude N, Frambes N, Brennen H, Robertson B, Hollis F. Mitochondrial might: powering the peripartum for risk and resilience. Front Behav Neurosci 2023; 17:1286811. [PMID: 38187925 PMCID: PMC10767224 DOI: 10.3389/fnbeh.2023.1286811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/01/2023] [Indexed: 01/09/2024] Open
Abstract
The peripartum period, characterized by dynamic hormonal shifts and physiological adaptations, has been recognized as a potentially vulnerable period for the development of mood disorders such as postpartum depression (PPD). Stress is a well-established risk factor for developing PPD and is known to modulate mitochondrial function. While primarily known for their role in energy production, mitochondria also influence processes such as stress regulation, steroid hormone synthesis, glucocorticoid response, GABA metabolism, and immune modulation - all of which are crucial for healthy pregnancy and relevant to PPD pathology. While mitochondrial function has been implicated in other psychiatric illnesses, its role in peripartum stress and mental health remains largely unexplored, especially in relation to the brain. In this review, we first provide an overview of mitochondrial involvement in processes implicated in peripartum mood disorders, underscoring their potential role in mediating pathology. We then discuss clinical and preclinical studies of mitochondria in the context of peripartum stress and mental health, emphasizing the need for better understanding of this relationship. Finally, we propose mitochondria as biological mediators of resilience to peripartum mood disorders.
Collapse
Affiliation(s)
- Erin Gorman-Sandler
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Healthcare System, Columbia, SC, United States
| | - Gabrielle Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Nazharee Cloude
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Noelle Frambes
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Hannah Brennen
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Breanna Robertson
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Healthcare System, Columbia, SC, United States
- USC Institute for Cardiovascular Disease Research, Columbia, SC, United States
| |
Collapse
|
4
|
Takenawa S, Nagasawa Y, Go K, Chérasse Y, Mizuno S, Sano K, Ogawa S. Activity of estrogen receptor β expressing neurons in the medial amygdala regulates preference toward receptive females in male mice. Proc Natl Acad Sci U S A 2023; 120:e2305950120. [PMID: 37819977 PMCID: PMC10589649 DOI: 10.1073/pnas.2305950120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/06/2023] [Indexed: 10/13/2023] Open
Abstract
The processing of information regarding the sex and reproductive state of conspecific individuals is critical for successful reproduction and survival in males. Generally, male mice exhibit a preference toward the odor of sexually receptive (RF) over nonreceptive females (XF) or gonadally intact males (IM). Previous studies suggested the involvement of estrogen receptor beta (ERβ) expressed in the medial amygdala (MeA) in male preference toward RF. To further delineate the role played by ERβ in the MeA in the neuronal network regulating male preference, we developed a new ERβ-iCre mouse line using the CRISPR-Cas9 system. Fiber photometry Ca2+ imaging revealed that ERβ-expressing neurons in the postero-dorsal part of the MeA (MeApd-ERβ+ neurons) were more active during social investigation toward RF compared to copresented XF or IM mice in a preference test. Chemogenetic inhibition of MeApd-ERβ+ neuronal activity abolished a preference to RF in "RF vs. XF," but not "RF vs. IM," tests. Analysis with cre-dependent retrograde tracing viral vectors identified the principal part of the bed nucleus of stria terminalis (BNSTp) as a primary projection site of MeApd-ERβ+ neurons. Fiber photometry recording in the BNSTp during a preference test revealed that chemogenetic inhibition of MeApd-ERβ+ neurons abolished differential neuronal activity of BNSTp cells as well as a preference to RF against XF but not against IM mice. Collectively, these findings demonstrate for the first time that MeApd-ERβ+ neuronal activity is required for expression of receptivity-based preference (i.e., RF vs. XF) but not sex-based preference (i.e., RF vs. IM) in male mice.
Collapse
Affiliation(s)
- Satoshi Takenawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Yutaro Nagasawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Kim Go
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Yoan Chérasse
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Trans-border Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba305-8575, Japan
| | - Kazuhiro Sano
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, Faculty of Human Sciences, University of Tsukuba, Tsukuba305-8577, Japan
| |
Collapse
|
5
|
Ronchetti S, Labombarda F, Roig P, De Nicola AF, Pietranera L. Beneficial effects of the phytoestrogen genistein on hippocampal impairments of spontaneously hypertensive rats (SHR). J Neuroendocrinol 2023; 35:e13228. [PMID: 36690381 DOI: 10.1111/jne.13228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/09/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023]
Abstract
Hippocampal neuropathology is a recognized feature of the spontaneously hypertensive rat (SHR). The hippocampal alterations associate with cognitive impairment. We have shown that hippocampal abnormalities are reversed by 17β-estradiol, a steroid binding to intracellular receptors (estrogen receptor α and β subtypes) or the membrane-located G-protein coupled estradiol receptor. Genistein (GEN) is a neuroprotective phytoestrogen which binds to estrogen receptor β and G-protein coupled estradiol receptor. Here, we investigated whether GEN neuroprotection extends to SHR. For this purpose, we treated 5-month-old SHR for 2 weeks with 10 mg kg-1 daily s.c injections of GEN. We analyzed the expression of doublecortin+ neuronal progenitors, glial fibrillary acidic protein+ astrocytes and ionized calcium-binding adapter molecule 1+ microglia in the CA1 region and dentate gyrus of the hippocampus using immunocytochemistry, whereas a quantitative real-time polymerase chain reaction was used to measure the expression of pro- and anti-inflammatory factors tumor necrosis factor α, cyclooxygenase-2 and transforming growth factor β. We also evaluated hippocampal dependent memory using the novel object recognition test. The results showed a decreased number of doublecortin+ neural progenitors in the dentate gyrus of SHR that was reversed with GEN. The number of glial fibrillary acidic protein+ astrocytes in the dentate gyrus and CA1 was increased in SHR but significantly decreased by GEN treatment. Additionally, GEN shifted microglial morphology from the predominantly activated phenotype present in SHR, to the more surveillance phenotype found in normotensive rats. Furthermore, treatment with GEN decreased the mRNA of the pro-inflammatory factors tumor necrosis factor α and cyclooxygenase-2 and increased the mRNA of the anti-inflammatory factor transforming growth factor β. Discrimination index in the novel object recognition test was decreased in SHR and treatment with GEN increased this parameter. Our results indicate important neuroprotective effects of GEN at the neurochemical and behavioral level in SHR. Our data open an interesting possibility for proposing this phytoestrogen as an alternative therapy in hypertensive encephalopathy.
Collapse
Affiliation(s)
- Santiago Ronchetti
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Florencia Labombarda
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Luciana Pietranera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
6
|
Biason-Lauber A, Lang-Muritano M. Estrogens: Two nuclear receptors, multiple possibilities. Mol Cell Endocrinol 2022; 554:111710. [PMID: 35787463 DOI: 10.1016/j.mce.2022.111710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022]
Abstract
Much is known about estrogen action in experimental animal models and in human physiology. This article reviews the mechanisms of estrogen activity in animals and humans and the role of its two receptors α and β in terms of structure and mechanisms of action in various tissues in health and in relationship with human pathologies (e.g., osteoporosis). Recently, the spectrum of clinical pictures of estrogen resistance caused by estrogen receptors gene variants has been widened by our description of a woman with β-receptor defect, which could be added to the already known descriptions of α-receptor defect in women and men and β-receptor defect in men. The essential role of the β-receptor in the development of the gonad stands out. We summarize the clinical pictures due to estrogen resistance in men and women and focus on long-term follow-up of two women, one with α- and the other with β-receptor resistance. Some open questions remain on the complex interactions between the two receptors on bone metabolism and hypothalamus-pituitary-gonadal axis, which need further deepening and research.
Collapse
Affiliation(s)
- Anna Biason-Lauber
- University of Fribourg, Division of Endocrinology, Chemin du Musée 5, 1700, Fribourg, Switzerland.
| | - Mariarosaria Lang-Muritano
- Division of Pediatric Endocrinology and Diabetology, Switzerland; Children's Research Center, University Children's Hospital, Steinwiesstrasse 75, 8032, Zurich, Switzerland
| |
Collapse
|
7
|
Fukui K, Sato K, Murakawa S, Minami M, Amano T. Estrogen signaling modulates behavioral selection toward pups and amygdalohippocampal area in the rhomboid nucleus of the bed nucleus of the stria terminalis circuit. Neuropharmacology 2022; 204:108879. [PMID: 34785164 DOI: 10.1016/j.neuropharm.2021.108879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023]
Abstract
Gonadal steroid hormone influences behavioral choice of adult animals toward pups, parental or aggressive. We previously reported that long-term administration of 17β-estradiol (E2) to male mice during sexual maturation induces aggressive behavior toward conspecific pups, which is called "infanticide," and significantly enhanced excitatory synaptic transmission in the rhomboid nucleus of bed nucleus of the stria terminalis (BSTrh), which is an important brain region for infanticide. However, it is unclear how estrogen receptor-dependent signaling after sexual maturity regulates neural circuits including the BSTrh. Here we revealed that E2 administration to gonadectomized mice in adulthood elicited infanticidal behavior and enhanced excitatory synaptic transmission in the BSTrh by increasing the probability of glutamate release from the presynaptic terminalis. Next, we performed whole-brain mapping of E2-sensitive brain regions projecting to the BSTrh and found that amygdalohippocampal area (AHi) neurons that project to the BSTrh densely express estrogen receptor 1 (Esr1). Moreover, E2 treatment enhanced synaptic connectivity in the AHi-BSTrh pathway. Together, these results suggest that reinforcement of excitatory inputs from AHi neurons into the BSTrh by estrogen receptor-dependent signaling may contribute to the expression of infanticide.
Collapse
Affiliation(s)
- Kiyoshiro Fukui
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Keiichiro Sato
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Shunsaku Murakawa
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Masabumi Minami
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan
| | - Taiju Amano
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, 060-0812, Japan.
| |
Collapse
|
8
|
Rainville JR, Lipuma T, Hodes GE. Translating the Transcriptome: Sex Differences in the Mechanisms of Depression and Stress, Revisited. Biol Psychiatry 2022; 91:25-35. [PMID: 33865609 PMCID: PMC10197090 DOI: 10.1016/j.biopsych.2021.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 02/01/2021] [Accepted: 02/01/2021] [Indexed: 12/28/2022]
Abstract
The past decade has produced a plethora of studies examining sex differences in the transcriptional profiles of stress and mood disorders. As we move forward from accepting the existence of extensive molecular sex differences in the brain to exploring the purpose of these sex differences, our approach must become more systemic and less reductionist. Earlier studies have examined specific brain regions and/or cell types. To use this knowledge to develop the next generation of personalized medicine, we need to comprehend how transcriptional changes across the brain and/or the body relate to each other. We provide an overview of the relationships between baseline and depression/stress-related transcriptional sex differences and explore contributions of preclinically identified mechanisms and their impacts on behavior.
Collapse
Affiliation(s)
- Jennifer R Rainville
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Timothy Lipuma
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Georgia E Hodes
- Department of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia.
| |
Collapse
|
9
|
He FQ, Fan MY, Hui YN, Lai RJ, Chen X, Yang MJ, Cheng XX, Wang ZJ, Yu B, Yan BJ, Tian Z. Effects of treadmill exercise on anxiety-like behavior in association with changes in estrogen receptors ERα, ERβ and oxytocin of C57BL/6J female mice. IBRO Neurosci Rep 2021; 11:164-174. [PMID: 34746914 PMCID: PMC8551837 DOI: 10.1016/j.ibneur.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/04/2021] [Indexed: 11/28/2022] Open
Abstract
Exercise can reduce the incidence of stress-related mental diseases, such as depression and anxiety. Control group was neither exposed to CVMS nor TRE (noCVMS/noTRE). Females were tested and levels of serum17-beta-oestradiol (E2), estrogen receptors α immunoreactive neurons (ERα-IRs), estrogen receptors β immunoreactive neurons (ERβ-IRs) and oxytocin immunoreactive neurons (OT-IRs) were measured. The results showed there's increased anxiety-like behaviors for mice from CVMS/noTRE, CVMS/higher speed TRE (CVMS/HTRE) and noCVMS/HTRE groups when they were put in open field and elevated maze tests. They had lower serum E2 levels than mice from CVMS/low-moderate speed TRE (CVMS/LMTRE), noCVMS/LMTRE and noCVMS/noTRE groups. The three groups of CVMS/noTRE, CVMS/HTRE and noCVMS/HTRE mice had more ERα-IRs and less ERβ-IRs in the medial preoptic area (mPOA), bed nucleus of the stria terminalis (BNST) and medial amygdala (MeA), hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON). The number of OT-IRs in PVN and SON of CVMS/noTRE, CVMS/HTRE and noCVMS/HTRE mice was also lower than that of mice from CVMS/LMTRE, noCVMS/LMTRE and noCVMS/noTRE groups. Interestingly, CVMS/LMTRE and noCVMS/LMTRE mice were similar to noCVMS/noTRE mice in that they did not show anxiety, while CVMS/HTRE and noCVMS/HTRE mice did not, which were similar to the mice in CVMS/noTRE. We propose that LMTRE instead of HTRE changes the serum concentration of E2. ERβ/ERα ratio and OT level in the brain may be responsible for the decrease in anxiety-like behavior in female mice exposed to anxiety-inducing stress conditions.
Collapse
Key Words
- BNST, bed nucleus of the stria terminalis
- CVMS, chronic variable moderate stress
- Chronic variable moderate stress (CVMS)
- E2, 17-beta-oestradiol
- ELISA, enzyme-linked immunosorbent assay
- EPM, elevated plusmazetest
- ERα-IRs, estrogen receptors αimmunoreactive neurons
- ERβ-IRs, estrogen receptor β immunoreactive neurons
- Estrogen receptor α (ERα)
- Estrogen receptor β (ERβ)
- HPA, hypothalamic–pituitary–adrenal
- HRP, horseradishperoxidase
- HTRE, higher speed TRE
- LMTRE, low-moderate speed TRE
- MeA, medial amygdaloid nucleus
- OF, open field test
- OT-IRs, Oxytocin immunoreactive neurons
- Oxytocin (OT)
- PBS, phosphatebufferedsolution
- PVN, paraventricular nucleus
- SON, supraoptic nucleus
- TRE, treadmill exercise
- Treadmill exercise (TRE)
- mPOA, medial preopticarea
Collapse
Affiliation(s)
- Feng-Qin He
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Mei-Yang Fan
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Yu-Nan Hui
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Rui-Juan Lai
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Xin Chen
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Ming-Juan Yang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Xiao-Xia Cheng
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Zi-Jian Wang
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Bin Yu
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Bing-Jie Yan
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| | - Zhen Tian
- Key Laboratory of Natural Product Development and Anticancer Innovative Drug Research in Qinling, Xi'an 710065, China.,Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an 710065, China
| |
Collapse
|
10
|
Kim CK, Linscott ML, Flury S, Zhang M, Newby ML, Pak TR. 17β-Estradiol Regulates miR-9-5p and miR-9-3p Stability and Function in the Aged Female Rat Brain. Noncoding RNA 2021; 7:53. [PMID: 34564315 PMCID: PMC8482090 DOI: 10.3390/ncrna7030053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
Clinical studies demonstrated that the ovarian hormone 17β-estradiol (E2) is neuroprotective within a narrow window of time following menopause, suggesting that there is a biological switch in E2 action that is temporally dependent. However, the molecular mechanisms mediating this temporal switch have not been determined. Our previous studies focused on microRNAs (miRNA) as one potential molecular mediator and showed that E2 differentially regulated a subset of mature miRNAs which was dependent on age and the length of time following E2 deprivation. Notably, E2 significantly increased both strands of the miR-9 duplex (miR-9-5p and miR-9-3p) in the hypothalamus, raising the possibility that E2 could regulate miRNA stability/degradation. We tested this hypothesis using a biochemical approach to measure miRNA decay in a hypothalamic neuronal cell line and in hypothalamic brain tissue from a rat model of surgical menopause. Notably, we found that E2 treatment stabilized both miRNAs in neuronal cells and in the rat hypothalamus. We also used polysome profiling as a proxy for miR-9-5p and miR-9-3p function and found that E2 was able to shift polysome loading of the miRNAs, which repressed the translation of a predicted miR-9-3p target. Moreover, miR-9-5p and miR-9-3p transcripts appeared to occupy different fractions of the polysome profile, indicating differential subcellular. localization. Together, these studies reveal a novel role for E2 in modulating mature miRNA behavior, independent of its effects at regulating the primary and/or precursor form of miRNAs.
Collapse
Affiliation(s)
| | | | | | | | | | - Toni R. Pak
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, IL 60153, USA; (C.K.K.); (M.L.L.); (S.F.); (M.Z.); (M.L.N.)
| |
Collapse
|
11
|
Amiresmaili S, Shahrokhi N, Khaksari M, AsadiKaram G, Aflatoonian MR, Shirazpour S, Amirkhosravi L, Mortazaeizadeh A. The Hepatoprotective mechanisms of 17β-estradiol after traumatic brain injury in male rats: Classical and non-classical estrogen receptors. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 213:111987. [PMID: 33582408 DOI: 10.1016/j.ecoenv.2021.111987] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 06/12/2023]
Abstract
Protective effects of estrogen (E2) on traumatic brain injury (TBI) have been determined. In this study, the hepatoprotective effects of E2 after TBI through its receptors and oxidative stress regulation have been evaluated. Diffuse TBI induced by the Marmarou method in male rats. G15, PHTPP, MPP, and ICI182-780 as selective antagonists of E2 were injected before TBI. The results indicated that TBI induces a significant increase in liver enzymes [Alkaline phosphatase (ALP), Aspartate aminotransferase (AST), Alanine aminotransferase (ALT), Glutamyl transferase (GGT)], and oxidants levels [Malondialdehyde (MDA), Nitric oxide (NO)] and decreases in antioxidant biomarkers [Glutathione peroxidase (GPx) and Superoxide dismutase (SOD)] in the brain and liver, and plasma. We also found that E2 significantly preserved levels of these biomarkers and enzymatic activity. All antagonists inhibited the effects of E2 on increasing SOD and GPx. Also, the effects of E2 on brain MDA levels were inhibited by all antagonists, but in the liver, only ICI + G15 + E2 + TBI group was affected. The impacts of E2 on brain and liver and plasma NO levels were inhibited by all antagonists. The current findings demonstrated that E2 probably improved liver injury after TBI by modulating oxidative stress. Also, both classic (ERβ, ERα) and non-classic [G protein-coupled estrogen receptor (GPER)] receptors are affected in the protective effects of E2.
Collapse
Affiliation(s)
- Sedigheh Amiresmaili
- Department of Physiology, Bam University of Medical Sciences, Bam, Iran; Physiology Research Center, Institute of Basic and Clinical Physiology Science, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Science, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza AsadiKaram
- Physiology Research Center, Institute of Basic and Clinical Physiology Science, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Shirazpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ladan Amirkhosravi
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Science, Kerman University of Medical Sciences, Kerman, Iran
| | - Abbas Mortazaeizadeh
- Researcher, Pathology and Stem Cells Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
12
|
Rigney N, Whylings J, de Vries GJ, Petrulis A. Sex Differences in the Control of Social Investigation and Anxiety by Vasopressin Cells of the Paraventricular Nucleus of the Hypothalamus. Neuroendocrinology 2021; 111:521-535. [PMID: 32541145 PMCID: PMC7736187 DOI: 10.1159/000509421] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
The neuropeptide arginine-vasopressin (AVP) has long been implicated in the regulation of social behavior and communication in diverse taxa, but the source of AVP release relevant for behavior has not been precisely determined. Potential sources include hypothalamic cell populations such as the paraventricular (PVN), supraoptic, and suprachiasmatic nuclei, as well as extrahypothalamic cell groups in the extended amygdala. To address if AVP-expressing cells in the PVN are important for mouse social communication, we deleted PVN AVP-expressing cells using viral-mediated delivery of Cre-dependent caspase-9 cell death construct into the PVN of AVP-Cre-positive mice (expressing Cre-recombinase under the control of the AVP promoter) or AVP-Cre-negative littermate controls, and assessed their levels of social investigation, social communication, anxiety, sex behavior, and aggressive behavior. We found that these lesions increased social investigation in females, but not in males. However, in males but not in females, these lesions increased non-social anxiety-related behaviors in the elevated-plus maze. These results therefore point at differential involvement of PVN AVP-expressing cells in the context of social and emotional behavior in the two sexes, which may contribute to sex differences in social communication and anxiety disorders.
Collapse
Affiliation(s)
- Nicole Rigney
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA,
| | - Jack Whylings
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Geert J de Vries
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Aras Petrulis
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Abi-Ghanem C, Robison LS, Zuloaga KL. Androgens' effects on cerebrovascular function in health and disease. Biol Sex Differ 2020; 11:35. [PMID: 32605602 PMCID: PMC7328272 DOI: 10.1186/s13293-020-00309-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Androgens affect the cerebral vasculature and may contribute to sex differences in cerebrovascular diseases. Men are at a greater risk for stroke and vascular contributions to cognitive impairment and dementia (VCID) compared to women throughout much of the lifespan. The cerebral vasculature is a target for direct androgen actions, as it expresses several sex steroid receptors and metabolizing enzymes. Androgens’ actions on the cerebral vasculature are complex, as they have been shown to have both protective and detrimental effects, depending on factors such as age, dose, and disease state. When administered chronically, androgens are shown to be pro-angiogenic, promote vasoconstriction, and influence blood-brain barrier permeability. In addition to these direct effects of androgens on the cerebral vasculature, androgens also influence other vascular risk factors that may contribute to sex differences in cerebrovascular diseases. In men, low androgen levels have been linked to metabolic and cardiovascular diseases including hypertension, diabetes, hyperlipidemia, and obesity, which greatly increase the risk of stroke and VCID. Thus, a better understanding of androgens’ interactions with the cerebral vasculature under physiological and pathological conditions is of key importance.
Collapse
Affiliation(s)
- Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
14
|
Neurochemical Characterization of Neurons Expressing Estrogen Receptor β in the Hypothalamic Nuclei of Rats Using in Situ Hybridization and Immunofluorescence. Int J Mol Sci 2019; 21:ijms21010115. [PMID: 31877966 PMCID: PMC6981915 DOI: 10.3390/ijms21010115] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/14/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Estrogens play an essential role in multiple physiological functions in the brain, including reproductive neuroendocrine, learning and memory, and anxiety-related behaviors. To determine these estrogen functions, many studies have tried to characterize neurons expressing estrogen receptors known as ERα and ERβ. However, the characteristics of ERβ-expressing neurons in the rat brain still remain poorly understood compared to that of ERα-expressing neurons. The main aim of this study is to determine the neurochemical characteristics of ERβ-expressing neurons in the rat hypothalamus using RNAscope in situ hybridization (ISH) combined with immunofluorescence. Strong Esr2 signals were observed especially in the anteroventral periventricular nucleus (AVPV), bed nucleus of stria terminalis, hypothalamic paraventricular nucleus (PVN), supraoptic nucleus, and medial amygdala, as previously reported. RNAscope ISH with immunofluorescence revealed that more than half of kisspeptin neurons in female AVPV expressed Esr2, whereas few kisspeptin neurons were found to co-express Esr2 in the arcuate nucleus. In the PVN, we observed a high ratio of Esr2 co-expression in arginine-vasopressin neurons and a low ratio in oxytocin and corticotropin-releasing factor neurons. The detailed neurochemical characteristics of ERβ-expressing neurons identified in the current study can be very essential to understand the estrogen signaling via ERβ.
Collapse
|
15
|
Activation of the G Protein-Coupled Estrogen Receptor (GPER) Increases Neurogenesis and Ameliorates Neuroinflammation in the Hippocampus of Male Spontaneously Hypertensive Rats. Cell Mol Neurobiol 2019; 40:711-723. [DOI: 10.1007/s10571-019-00766-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/22/2019] [Indexed: 01/20/2023]
|
16
|
Balla B, Sárvári M, Kósa JP, Kocsis-Deák B, Tobiás B, Árvai K, Takács I, Podani J, Liposits Z, Lakatos P. Long-term selective estrogen receptor-beta agonist treatment modulates gene expression in bone and bone marrow of ovariectomized rats. J Steroid Biochem Mol Biol 2019; 188:185-194. [PMID: 30685384 DOI: 10.1016/j.jsbmb.2019.01.012] [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: 07/10/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 01/20/2023]
Abstract
Gonadal hormones including 17β-estradiol exert important protective functions in skeletal homeostasis. However, numerous details of ovarian hormone deficiency in the common bone marrow microenvironment have not yet been revealed and little information is available on the tissue-specific acts either, especially those via estrogen receptor beta (ERβ). The aim of the present study was therefore to examine the bone-related gene expression changes after ovariectomy (OVX) and long-term ERβ agonist diarylpropionitrile (DPN) administration. We found that OVX produced strong and widespread changes of gene expression in both femoral bone and bone marrow. In the bone out of 22 genes, 20 genes were up- and 2 were downregulated after OVX. It is noteworthy that DPN restored mRNA expression of 10 OVX-induced changes (Aldh2, Col1a1, Daam1, Fgf12, Igf1, Il6r, Nfkb1, Notch1, Notch2 and Psen1) suggesting a modulatory role of ERβ in bone physiology. In bone marrow, out of 37 categorized genes, transcription of 25 genes were up- and 12 were downregulated indicating that the marrow is highly responsive to gonadal hormones. DPN modestly affected transcription, only expression of two genes (Nfatc1 and Tgfb1) was restored by DPN action. The PI3K/Akt signaling pathway was the most affected gene cluster following the interventions in bone and bone marrow, as demonstrated by canonical variates analysis (CVA). We suggested that our results contribute to a deeper understanding of alterations in gene expression of bone and bone marrow niche elicited by ERβ and selective ERβ analogs.
Collapse
Affiliation(s)
- Bernadett Balla
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary.
| | - Miklós Sárvári
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - János P Kósa
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Barbara Kocsis-Deák
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Bálint Tobiás
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Kristóf Árvai
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - István Takács
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - János Podani
- Biological Institute, Eötvös Loránd University, Budapest, Hungary
| | - Zsolt Liposits
- Laboratory of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary; Department of Neuroscience, Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Péter Lakatos
- 1st Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
17
|
Nakata M, Ågmo A, Sagoshi S, Ogawa S. The Role of Estrogen Receptor β (ERβ) in the Establishment of Hierarchical Social Relationships in Male Mice. Front Behav Neurosci 2018; 12:245. [PMID: 30405370 PMCID: PMC6204783 DOI: 10.3389/fnbeh.2018.00245] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 10/01/2018] [Indexed: 11/13/2022] Open
Abstract
Acquisition of social dominance is important for social species including mice, for preferential access to foods and mates. Male mice establish social rank through agonistic behaviors, which are regulated by gonadal steroid hormone, testosterone, as its original form and aromatized form. It is well known that estrogen receptors (ERs), particularly ER α (ERα), mediate effects of aromatized testosterone, i.e., 17β-estradiol, but precise role played by ER β (ERβ) is still unclear. In the present study, we investigated effects of ERβ gene disruption on social rank establishment in male mice. Adult male ERβ knockout (βERKO) mice and their wild type (WT) littermates were paired based on genotype- and weight-matched manner and tested against each other repeatedly during 7 days experimental period. They underwent 4 trials of social interaction test in neutral cage (homogeneous set test) every other day. Along repeated trials, WT but not βERKO pairs showed a gradual increase of agonistic behaviors including aggression and tail rattling, and a gradual decrease of latency to social rank determination in tube test conducted after each trial of the social interaction test. Analysis of behavioral transition further suggested that WT winners in the tube test showed one-sided aggression during social interaction test suggesting WT pairs went through a process of social rank establishment. On the other hand, a dominant-subordinate relationship in βERKO pairs was not as apparent as that in WT pairs. Moreover, βERKO mice showed lower levels of aggressive behavior than WT mice in social interaction tests. These findings collectively suggest that ERβ may play a significant role in the establishment and maintenance of hierarchical social relationships among male mice.
Collapse
Affiliation(s)
- Mariko Nakata
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan.,Research Fellow, Japan Society for Promotion of Science (JSPS), Tokyo, Japan
| | - Anders Ågmo
- Department of Psychology, University of Tromsø, Tromsø, Norway
| | - Shoko Sagoshi
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
18
|
Lang-Muritano M, Sproll P, Wyss S, Kolly A, Hürlimann R, Konrad D, Biason-Lauber A. Early-Onset Complete Ovarian Failure and Lack of Puberty in a Woman With Mutated Estrogen Receptor β (ESR2). J Clin Endocrinol Metab 2018; 103:3748-3756. [PMID: 30113650 DOI: 10.1210/jc.2018-00769] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/27/2018] [Indexed: 02/04/2023]
Abstract
CONTEXT Estrogen resistance due to mutations in the estrogen receptor α gene (ESR1) has been described in men and women and is characterized by osteoporosis, delayed bone age and continuous growth in adulthood, and delayed puberty and multiple ovarian cysts in women. Although mutations in the estrogen receptor β gene ESR2 were found in 46, XY patients with differences of sex development, no genetic variants of ESR2 were linked to gonadal defects in women. SETTINGS AND PATIENT Here we describe a 16-year-old female patient who came to our tertiary care hospital with complete lack of estrogen action, as demonstrated by absent breast development, primary amenorrhea, and osteoporosis, resembling patients with ESR1 mutation. However, her gonads were clearly abnormal (streak), a finding not observed in ESR1-deficient patients. DESIGN To gain insights into the molecular consequences of the ESR2 defect, whole exome sequencing and extensive functional transactivation studies in ovarian, bone, and breast cells were conducted, with or without the natural activator of estrogen receptors, 17β-estradiol. RESULTS We identified a loss-of-function heterozygous mutation of a highly conserved residue in ESR2 that disrupts estradiol-dependent signaling and has a dominant negative effect, most likely due to failure to interact with its coactivator, nuclear coactivator 1. CONCLUSIONS This is a report of a loss-of-function mutation in the estrogen receptor β in a young woman with complete ovarian failure, suggesting that ESR2 is necessary for human ovarian determination and/or maintenance and that ESR1 is not sufficient to sustain ovarian function in humans.
Collapse
Affiliation(s)
- Mariarosaria Lang-Muritano
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Patrick Sproll
- Division of Endocrinology, University of Fribourg, Fribourg, Switzerland
| | - Sascha Wyss
- Division of Endocrinology, University of Fribourg, Fribourg, Switzerland
| | - Anne Kolly
- Division of Endocrinology, University of Fribourg, Fribourg, Switzerland
| | - Renate Hürlimann
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland
- Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Anna Biason-Lauber
- Division of Endocrinology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
19
|
Zhang R, Zhang Y, Wu M, Yan P, Izaz A, Wang R, Zhu H, Zhou Y, Wu X. Molecular cloning of androgen receptor and gene expression of sex steroid hormone receptors in the brain of newborn Chinese alligator (Alligator sinensis). Gene 2018; 674:178-187. [DOI: 10.1016/j.gene.2018.06.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/29/2018] [Accepted: 06/11/2018] [Indexed: 12/16/2022]
|
20
|
Krolick KN, Zhu Q, Shi H. Effects of Estrogens on Central Nervous System Neurotransmission: Implications for Sex Differences in Mental Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:105-171. [PMID: 30470289 PMCID: PMC6737530 DOI: 10.1016/bs.pmbts.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nearly one of every five US individuals aged 12 years old or older lives with certain types of mental disorders. Men are more likely to use various types of substances, while women tend to be more susceptible to mood disorders, addiction, and eating disorders, all of which are risks associated with suicidal attempts. Fundamental sex differences exist in multiple aspects of the functions and activities of neurotransmitter-mediated neural circuits in the central nervous system (CNS). Dysregulation of these neural circuits leads to various types of mental disorders. The potential mechanisms of sex differences in the CNS neural circuitry regulating mood, reward, and motivation are only beginning to be understood, although they have been largely attributed to the effects of sex hormones on CNS neurotransmission pathways. Understanding this topic is important for developing prevention and treatment of mental disorders that should be tailored differently for men and women. Studies using animal models have provided important insights into pathogenesis, mechanisms, and new therapeutic approaches of human diseases, but some concerns remain to be addressed. The purpose of this chapter is to integrate human and animal studies involving the effects of the sex hormones, estrogens, on CNS neurotransmission, reward processing, and associated mental disorders. We provide an overview of existing evidence for the physiological, behavioral, cellular, and molecular actions of estrogens in the context of controlling neurotransmission in the CNS circuits regulating mood, reward, and motivation and discuss related pathology that leads to mental disorders.
Collapse
Affiliation(s)
- Kristen N Krolick
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Qi Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States; Cellular, Molecular and Structural Biology, Miami University, Oxford, OH, United States.
| |
Collapse
|
21
|
Gourdy P, Guillaume M, Fontaine C, Adlanmerini M, Montagner A, Laurell H, Lenfant F, Arnal JF. Estrogen receptor subcellular localization and cardiometabolism. Mol Metab 2018; 15:56-69. [PMID: 29807870 PMCID: PMC6066739 DOI: 10.1016/j.molmet.2018.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In addition to their crucial role in reproduction, estrogens are key regulators of energy and glucose homeostasis and they also exert several cardiovascular protective effects. These beneficial actions are mainly mediated by estrogen receptor alpha (ERα), which is widely expressed in metabolic and vascular tissues. As a member of the nuclear receptor superfamily, ERα was primarily considered as a transcription factor that controls gene expression through the activation of its two activation functions (ERαAF-1 and ERαAF-2). However, besides these nuclear actions, a pool of ERα is localized in the vicinity of the plasma membrane, where it mediates rapid signaling effects called membrane-initiated steroid signals (MISS) that have been well described in vitro, especially in endothelial cells. SCOPE OF THE REVIEW This review aims to summarize our current knowledge of the mechanisms of nuclear vs membrane ERα activation that contribute to the cardiometabolic protection conferred by estrogens. Indeed, new transgenic mouse models (affecting either DNA binding, activation functions or membrane localization), together with the use of novel pharmacological tools that electively activate membrane ERα effects recently allowed to begin to unravel the different modes of ERα signaling in vivo. CONCLUSION Altogether, available data demonstrate the prominent role of ERα nuclear effects, and, more specifically, of ERαAF-2, in the preventive effects of estrogens against obesity, diabetes, and atheroma. However, membrane ERα signaling selectively mediates some of the estrogen endothelial/vascular effects (NO release, reendothelialization) and could also contribute to the regulation of energy balance, insulin sensitivity, and glucose metabolism. Such a dissection of ERα biological functions related to its subcellular localization will help to understand the mechanism of action of "old" ER modulators and to design new ones with an optimized benefit/risk profile.
Collapse
Affiliation(s)
- Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Toulouse, France.
| | - Maeva Guillaume
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service d'Hépatologie et Gastro-Entérologie, CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| |
Collapse
|
22
|
Ohtani N, Suda K, Tsuji E, Tanemura K, Yokota H, Inoue H, Iwano H. Late pregnancy is vulnerable period for exposure to BPA. J Vet Med Sci 2018; 80:536-543. [PMID: 29367495 PMCID: PMC5880839 DOI: 10.1292/jvms.17-0460] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Bisphenol A (BPA) is among the better-known endocrine disruptors. BPA is used in various food-contacting materials and is easily eluted into food; as a result, we are exposed to BPA on a daily basis. In adults, BPA is
metabolized and eliminated rapidly from the body. However, numerous reports suggest that fetuses and young children are susceptible to BPA. One of the concerning adverse effects of BPA is disruption of behavior,
especially anxiety-like behavior. In order to study the mechanism of influences on offspring, it is important to clarify the most vulnerable gestation period. We hypothesized that offspring in late pregnancy would be
more susceptible to BPA, because late pregnancy is a critical time for functional brain development. In this study, C57BL/6 mouse fetuses were exposed prenatally by oral dosing of pregnant dams, once daily from
gestational day 5.5 to 12.5 (early pregnancy) or 11.5 to 18.5 (late pregnancy), with BPA (0 or 10 mg/kg body weight). Following birth and weaning, the resulting pups were tested using an elevated plus maze at postnatal
week 10. The behavior of the offspring was altered by prenatal BPA exposure during late pregnancy but not during early pregnancy. These results indicated that offspring are more vulnerable to exposure to BPA in late
pregnancy.
Collapse
Affiliation(s)
- Naoko Ohtani
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Koshi Suda
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Erika Tsuji
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, Sendai, Miyagi 981-8555, Japan
| | - Hiroshi Yokota
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroki Inoue
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| | - Hidetomo Iwano
- Laboratory of Veterinary Biochemistry, Department of Bioscience, School of Veterinary Medicine, Rakuno Gakuen University, 582 Bunkyodai-Midorimachi, Ebetsu, Hokkaido 069-8501, Japan
| |
Collapse
|
23
|
Colón JM, González PA, Cajigas Á, Maldonado WI, Torrado AI, Santiago JM, Salgado IK, Miranda JD. Continuous tamoxifen delivery improves locomotor recovery 6h after spinal cord injury by neuronal and glial mechanisms in male rats. Exp Neurol 2018; 299:109-121. [PMID: 29037533 PMCID: PMC5723542 DOI: 10.1016/j.expneurol.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
No treatment is available for patients with spinal cord injury (SCI). Patients often arrive to the hospital hours after SCI suggesting the need of a therapy that can be used on a clinically relevant window. Previous studies showed that Tamoxifen (TAM) treatment 24h after SCI benefits locomotor recovery in female rats. Tamoxifen exerts beneficial effects in male and female rodents but a gap of knowledge exists on: the therapeutic window of TAM, the spatio-temporal mechanisms activated and if this response is sexually dimorphic. We hypothesized that TAM will favor locomotor recovery when administered up-to 24h after SCI in male Sprague-Dawley rats. Rats received a thoracic (T10) contusion using the MACSIS impactor followed by placebo or TAM (15mg/21days) pellets in a therapeutic window of 0, 6, 12, or 24h. Animals were sacrificed at 2, 7, 14, 28 or 35days post injury (DPI) to study the molecular and cellular changes in the acute and chronic stages. Immediate or delayed therapy (t=6h) improved locomotor function, increased white matter spared tissue, and neuronal survival. TAM reduced reactive gliosis during chronic stages and increased the expression of Olig-2. A significant difference was observed in estrogen receptor alpha between male and female rodents from 2 to 28 DPI suggesting a sexually dimorphic characteristic that could be related to the behavioral differences observed in the therapeutic window of TAM. This study supports the use of TAM in the SCI setting due to its neuroprotective effects but with a significant sexually dimorphic therapeutic window.
Collapse
Affiliation(s)
- Jennifer M Colón
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Pablo A González
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Ámbar Cajigas
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Wanda I Maldonado
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Aranza I Torrado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - José M Santiago
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Iris K Salgado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Jorge D Miranda
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| |
Collapse
|
24
|
Nowacka-Chmielewska MM, Kasprowska-Liśkiewicz D, Barski JJ, Obuchowicz E, Małecki A. The behavioral and molecular evaluation of effects of social instability stress as a model of stress-related disorders in adult female rats. Stress 2017; 20:549-561. [PMID: 28911267 DOI: 10.1080/10253890.2017.1376185] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The study aimed to test the hypotheses that chronic social instability stress (CSIS) alters behavioral and physiological parameters and expression of selected genes important for stress response and social behaviors. Adult female Sprague-Dawley rats were subjected to the 4-week CSIS procedure, which involves unpredictable rotation between phases of isolation and overcrowding. Behavioral analyses (Experiment 1) were performed on the same rats before and after CSIS (n = 16) and physiological and biochemical measurements (Experiment 2) were made on further control (CON; n = 7) and stressed groups (CSIS; n = 8). Behaviors in the open field test (locomotor and exploratory activities) and elevated-plus maze (anxiety-related behaviors) indicated anxiety after CSIS. CSIS did not alter the physiological parameters measured, i.e. body weight gain, regularity of estrous cycles, and circulating concentrations of stress hormones and sex steroids. QRT-PCR analysis of mRNA expression levels was performed on amygdala, hippocampus, prefrontal cortex (PFC), and hypothalamus. The main finding is that CSIS alters the mRNA levels for the studied genes in a region-specific manner. Hence, expression of POMC (pro-opiomelanocortin), AVPR1a (arginine vasopressin receptor), and OXTR (oxytocin receptor) significantly increased in the amygdala following CSIS, while in PFC and/or hypothalamus, POMC, AVPR1a, AVPR1b, OXTR, and ERβ (estrogen receptor beta) expression decreased. CSIS significantly reduced expression of CRH-R1 (corticotropin-releasing hormone receptor type 1) in the hippocampus. The directions of change in gene expression and the genes and regions affected indicate a molecular basis for the behavior changes. In conclusion, CSIS may be valuable for further analyzing the neurobiology of stress-related disorders in females.
Collapse
MESH Headings
- Amygdala/metabolism
- Animals
- Anxiety/genetics
- Anxiety/metabolism
- Behavior, Animal
- Brain/metabolism
- Chronic Disease
- Estrogen Receptor beta/genetics
- Estrogen Receptor beta/metabolism
- Female
- Gene Expression
- Hippocampus/metabolism
- Hypothalamo-Hypophyseal System/metabolism
- Hypothalamus/metabolism
- Pituitary-Adrenal System/metabolism
- Prefrontal Cortex/metabolism
- Pro-Opiomelanocortin/genetics
- Pro-Opiomelanocortin/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Receptors, Oxytocin/genetics
- Receptors, Oxytocin/metabolism
- Receptors, Vasopressin/genetics
- Receptors, Vasopressin/metabolism
- Stress, Psychological/genetics
- Stress, Psychological/metabolism
Collapse
Affiliation(s)
- Marta Maria Nowacka-Chmielewska
- a Laboratory of Molecular Biology, Faculty of Physiotherapy , The Jerzy Kukuczka Academy of Physical Education , Katowice , Poland
- b Department of Experimental Medicine, School of Medicine in Katowice , Medical University of Silesia , Katowice , Poland
| | - Daniela Kasprowska-Liśkiewicz
- a Laboratory of Molecular Biology, Faculty of Physiotherapy , The Jerzy Kukuczka Academy of Physical Education , Katowice , Poland
- b Department of Experimental Medicine, School of Medicine in Katowice , Medical University of Silesia , Katowice , Poland
| | - Jarosław Jerzy Barski
- b Department of Experimental Medicine, School of Medicine in Katowice , Medical University of Silesia , Katowice , Poland
- c Department of Physiology, School of Medicine in Katowice , Medical University of Silesia , Katowice , Poland
| | - Ewa Obuchowicz
- d Department of Pharmacology, School of Medicine in Katowice , Medical University of Silesia , Katowice , Poland
| | - Andrzej Małecki
- a Laboratory of Molecular Biology, Faculty of Physiotherapy , The Jerzy Kukuczka Academy of Physical Education , Katowice , Poland
| |
Collapse
|
25
|
Arambula SE, Jima D, Patisaul HB. Prenatal bisphenol A (BPA) exposure alters the transcriptome of the neonate rat amygdala in a sex-specific manner: a CLARITY-BPA consortium study. Neurotoxicology 2017; 65:207-220. [PMID: 29097150 DOI: 10.1016/j.neuro.2017.10.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Abstract
Bisphenol A (BPA) is a widely recognized endocrine disruptor prevalent in many household items. Because experimental and epidemiological data suggest links between prenatal BPA exposure and altered affective behaviors in children, even at levels below the current US FDA No Observed Adverse Effect Level (NOAEL) of 5mg/kg body weight (bw)/day, there is concern that early life exposure may alter neurodevelopment. The current study was conducted as part of the CLARITY-BPA (Consortium Linking Academic and Regulatory Insights on BPA Toxicity) program and examined the full amygdalar transcriptome on postnatal day (PND) 1, with the hypothesis that prenatal BPA exposure would alter the expression of genes and pathways fundamental to sex-specific affective behaviors. NCTR Sprague-Dawley dams were gavaged from gestational day 6 until parturition with BPA (2.5, 25, 250, 2500, or 25000μg/kg bw/day), a reference estrogen (0.05 or 0.5μg ethinyl estradiol (EE2)/kg bw/day), or vehicle. PND 1 amygdalae were microdissected and gene expression was assessed with qRT-PCR (all exposure groups) and RNAseq (vehicle, 25 and 250μg BPA, and 0.5μg EE2 groups only). Our results demonstrate that that prenatal BPA exposure can disrupt the transcriptome of the neonate amygdala, at doses below the FDA NOAEL, in a sex-specific manner and indicate that the female amygdala may be more sensitive to BPA exposure during fetal development. We also provide additional evidence that developmental BPA exposure can interfere with estrogen, oxytocin, and vasopressin signaling pathways in the developing brain and alter signaling pathways critical for synaptic organization and transmission.
Collapse
Affiliation(s)
- Sheryl E Arambula
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA
| | - Dereje Jima
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA; Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; WM Keck Center for Behavioral Biology, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
26
|
Maney DL. Polymorphisms in sex steroid receptors: From gene sequence to behavior. Front Neuroendocrinol 2017; 47:47-65. [PMID: 28705582 PMCID: PMC6312198 DOI: 10.1016/j.yfrne.2017.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/05/2017] [Accepted: 07/08/2017] [Indexed: 01/09/2023]
Abstract
Sex steroid receptors have received much interest as potential mediators of human behaviors and mental disorders. Candidate gene association studies have identified about 50 genetic variants of androgen and estrogen receptors that correlate with human behavioral phenotypes. Because most of these polymorphisms lie outside coding regions, discerning their effect on receptor function is not straightforward. Thus, although discoveries of associations improve our ability to predict risk, they have not greatly advanced our understanding of underlying mechanisms. This article is intended to serve as a starting point for psychologists and other behavioral biologists to consider potential mechanisms. Here, I review associations between polymorphisms in sex steroid receptors and human behavioral phenotypes. I then consider ways in which genetic variation can affect processes such as mRNA transcription, splicing, and stability. Finally, I suggest ways that hypotheses about mechanism can be tested, for example using in vitro assays and/or animal models.
Collapse
Affiliation(s)
- Donna L Maney
- Department of Psychology, 36 Eagle Row, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
27
|
Engler-Chiurazzi EB, Brown CM, Povroznik JM, Simpkins JW. Estrogens as neuroprotectants: Estrogenic actions in the context of cognitive aging and brain injury. Prog Neurobiol 2017; 157:188-211. [PMID: 26891883 PMCID: PMC4985492 DOI: 10.1016/j.pneurobio.2015.12.008] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/06/2015] [Accepted: 12/10/2015] [Indexed: 12/30/2022]
Abstract
There is ample empirical evidence to support the notion that the biological impacts of estrogen extend beyond the gonads to other bodily systems, including the brain and behavior. Converging preclinical findings have indicated a neuroprotective role for estrogen in a variety of experimental models of cognitive function and brain insult. However, the surprising null or even detrimental findings of several large clinical trials evaluating the ability of estrogen-containing hormone treatments to protect against age-related brain changes and insults, including cognitive aging and brain injury, led to hesitation by both clinicians and patients in the use of exogenous estrogenic treatments for nervous system outcomes. That estrogen-containing therapies are used by tens of millions of women for a variety of health-related applications across the lifespan has made identifying conditions under which benefits with estrogen treatment will be realized an important public health issue. Here we provide a summary of the biological actions of estrogen and estrogen-containing formulations in the context of aging, cognition, stroke, and traumatic brain injury. We have devoted special attention to highlighting the notion that estrogen appears to be a conditional neuroprotectant whose efficacy is modulated by several interacting factors. By developing criteria standards for desired beneficial peripheral and neuroprotective outcomes among unique patient populations, we can optimize estrogen treatments for attenuating the consequences of, and perhaps even preventing, cognitive aging and brain injury.
Collapse
Affiliation(s)
- E B Engler-Chiurazzi
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| | - C M Brown
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Neurobiology and Anatomy, West Virginia University, Morgantown, WV 26506, United States.
| | - J M Povroznik
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Pediatrics, West Virginia University, Morgantown, WV 26506, United States.
| | - J W Simpkins
- Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, WV 26506, United States; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
28
|
Oyola MG, Handa RJ. Hypothalamic-pituitary-adrenal and hypothalamic-pituitary-gonadal axes: sex differences in regulation of stress responsivity. Stress 2017; 20:476-494. [PMID: 28859530 PMCID: PMC5815295 DOI: 10.1080/10253890.2017.1369523] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gonadal hormones play a key role in the establishment, activation, and regulation of the hypothalamic-pituitary-adrenal (HPA) axis. By influencing the response and sensitivity to releasing factors, neurotransmitters, and hormones, gonadal steroids help orchestrate the gain of the HPA axis to fine-tune the levels of stress hormones in the general circulation. From early life to adulthood, gonadal steroids can differentially affect the HPA axis, resulting in sex differences in the responsivity of this axis. The HPA axis influences many physiological functions making an organism's response to changes in the environment appropriate for its reproductive status. Although the acute HPA response to stressors is a beneficial response, constant activation of this circuitry by chronic or traumatic stressful episodes may lead to a dysregulation of the HPA axis and cause pathology. Compared to males, female mice and rats show a more robust HPA axis response, as a result of circulating estradiol levels which elevate stress hormone levels during non-threatening situations, and during and after stressors. Fluctuating levels of gonadal steroids in females across the estrous cycle are a major factor contributing to sex differences in the robustness of HPA activity in females compared to males. Moreover, gonadal steroids may also contribute to epigenetic and organizational influences on the HPA axis even before puberty. Correspondingly, crosstalk between the hypothalamic-pituitary-gonadal (HPG) and HPA axes could lead to abnormalities of stress responses. In humans, a dysregulated stress response is one of the most common symptoms seen across many neuropsychiatric disorders, and as a result, such interactions may exacerbate peripheral pathologies. In this review, we discuss the HPA and HPG axes and review how gonadal steroids interact with the HPA axis to regulate the stress circuitry during all stages in life.
Collapse
Affiliation(s)
- Mario G Oyola
- a Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| | - Robert J Handa
- a Department of Biomedical Sciences , Colorado State University , Fort Collins , CO , USA
| |
Collapse
|
29
|
Molecular cloning of ESR2 and gene expression analysis of ESR1 and ESR2 in the pituitary gland of the Chinese alligator ( Alligator sinensis ) during female reproductive cycle. Gene 2017; 623:15-23. [DOI: 10.1016/j.gene.2017.04.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/04/2017] [Accepted: 04/12/2017] [Indexed: 02/02/2023]
|
30
|
Arnal JF, Lenfant F, Metivier R, Flouriot G, Henrion D, Adlanmerini M, Fontaine C, Gourdy P, Chambon P, Katzenellenbogen B, Katzenellenbogen J. Membrane and Nuclear Estrogen Receptor Alpha Actions: From Tissue Specificity to Medical Implications. Physiol Rev 2017; 97:1045-1087. [DOI: 10.1152/physrev.00024.2016] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/19/2016] [Accepted: 01/18/2017] [Indexed: 12/22/2022] Open
Abstract
Estrogen receptor alpha (ERα) has been recognized now for several decades as playing a key role in reproduction and exerting functions in numerous nonreproductive tissues. In this review, we attempt to summarize the in vitro studies that are the basis of our current understanding of the mechanisms of action of ERα as a nuclear receptor and the key roles played by its two activation functions (AFs) in its transcriptional activities. We then depict the consequences of the selective inactivation of these AFs in mouse models, focusing on the prominent roles played by ERα in the reproductive tract and in the vascular system. Evidence has accumulated over the two last decades that ERα is also associated with the plasma membrane and activates non-nuclear signaling from this site. These rapid/nongenomic/membrane-initiated steroid signals (MISS) have been characterized in a variety of cell lines, and in particular in endothelial cells. The development of selective pharmacological tools that specifically activate MISS and the generation of mice expressing an ERα protein impeded for membrane localization have begun to unravel the physiological role of MISS in vivo. Finally, we discuss novel perspectives for the design of tissue-selective ER modulators based on the integration of the physiological and pathophysiological roles of MISS actions of estrogens.
Collapse
Affiliation(s)
- Jean-Francois Arnal
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Françoise Lenfant
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Raphaël Metivier
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Gilles Flouriot
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Daniel Henrion
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Marine Adlanmerini
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Coralie Fontaine
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Gourdy
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Pierre Chambon
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - Benita Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| | - John Katzenellenbogen
- I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM) U 1048, Université de Toulouse 3 and CHU de Toulouse, Toulouse, France; Equipe SP@RTE UMR 6290 CNRS, Institut de Genétique et Développement de Rennes, Université de Rennes 1, Campus de Beaulieu, Rennes, France; Université de Rennes 1, Institut de Recherche en Santé, Environnement et Travail (Irest–INSERM UMR 1085), Equipe TREC, Rennes, France; Unité Mixte de Recherche 6214, Centre National de la Recherche Scientifique, Angers,
| |
Collapse
|
31
|
Patisaul HB. Endocrine disruption by dietary phyto-oestrogens: impact on dimorphic sexual systems and behaviours. Proc Nutr Soc 2017; 76:130-144. [PMID: 27389644 PMCID: PMC5646220 DOI: 10.1017/s0029665116000677] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A wide range of health benefits have been ascribed to soya intake including a lowered risk of osteoporosis, heart disease, breast cancer, and menopausal symptoms. Because it is a hormonally active diet, however, soya can also be endocrine disrupting, suggesting that intake has the potential to cause adverse health effects in certain circumstances, particularly when exposure occurs during development. Consequently, the question of whether or not soya phyto-oestrogens are beneficial or harmful to human health is neither straightforward nor universally applicable to all groups. Possible benefits and risks depend on age, health status, and even the presence or absence of specific gut microflora. As global consumption increases, greater awareness and consideration of the endocrine-disrupting properties of soya by nutrition specialists and other health practitioners is needed. Consumption by infants and small children is of particular concern because their hormone-sensitive organs, including the brain and reproductive system, are still undergoing sexual differentiation and maturation. Thus, their susceptibility to the endocrine-disrupting activities of soya phyto-oestrogens may be especially high. As oestrogen receptor partial agonists with molecular and cellular properties similar to anthropogenic endocrine disruptors such as bisphenol A, the soya phyto-oestrogens provide an interesting model for how attitudes about what is 'synthetic' v. what is 'natural,' shapes understanding and perception of what it means for a compound to be endocrine disrupting and/or potentially harmful. This review describes the endocrine-disrupting properties of soya phyto-oestrogens with a focus on neuroendocrine development and behaviour.
Collapse
Affiliation(s)
- Heather B Patisaul
- Department of Biological Sciences,Center for Human Health and the Environment,NC State University,Raleigh,NC 27695,USA
| |
Collapse
|
32
|
Wang X, Huang Y, Yuan S, Tamadon A, Ma S, Feng Y. The Role of Hippocampal Estradiol Receptor- α in a Perimenopausal Affective Disorders-Like Rat Model and Attenuating of Anxiety by Electroacupuncture. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:4958312. [PMID: 28044085 PMCID: PMC5156811 DOI: 10.1155/2016/4958312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/16/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Hormone replacement therapy is the principal treatment for perimenopausal affective disorders which can cause severe side effects. The present study compared the effects of electroacupuncture (EA) and estradiol treatment on perimenopausal affective disorders at the behavioral and cellular levels. In this randomized experimental in vivo study, adult female rats were divided into intact, ovariectomy, chronic unpredictable stress (CUS), and ovariectomy and CUS combination groups. After week 6, all groups were subdivided to three subgroups of control, EA, and estradiol treatment. The behavioral parameters in the open field and the elevated plus maze tests were assessed before and after treatments. Alterations of serum steroid hormones and changes of estradiol receptor-α (ER-α) immunofluorescence neurons in the hippocampus sections were evaluated. EA treatment caused more antianxiety effects than estradiol treatment in CUS group (P < 0.05). Notably, estradiol and EA treatments had better significant behavioral effects when the models were not estrogen-deficient. Importantly, within each group, compared to the control group, the numbers of ER-α-positive neurons were significantly larger in EA subgroups. Therefore, EA had antianxiety effects on perimenopausal affective disorders caused by CUS but not by estrogen deficiency and upregulation of hippocampus ER-α neurons may contribute to its mechanism of action.
Collapse
Affiliation(s)
- Xun Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongheng Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shiwen Yuan
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Amin Tamadon
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Shulan Ma
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
33
|
Pietranera L, Correa J, Brocca ME, Roig P, Lima A, Di Giorgio N, Garcia-Segura LM, De Nicola AF. Selective Oestrogen Receptor Agonists Rescued Hippocampus Parameters in Male Spontaneously Hypertensive Rats. J Neuroendocrinol 2016; 28. [PMID: 27517478 DOI: 10.1111/jne.12415] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 08/09/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022]
Abstract
Spontaneously hypertensive rats (SHR) show pronounced hippocampus alterations, including low brain-derived neurotrophic factor (BDNF) expression, reduced neurogenesis, astrogliosis and increased aromatase expression. These changes are reverted by treatment with 17β-oestradiol. To determine which oestradiol receptor (ER) type is involved in these neuroprotective effects, we used agonists of the ERα [propylpyrazole triol (PPT)] and the ERβ [diarylpropionitrite (DPN)] given over 2 weeks to 4-month-old male SHR. Wistar Kyoto normotensive rats served as controls. Using immunocytochemistry, we determined glial fibrillary protein (GFAP)+ astrocytes in the CA1, CA3 and hilus of the dentate gyrus of the hippocampus, aromatase immunostaining in the hilus, and doublecortin (DCX)+ neuronal progenitors in the inner granular zone of the dentate gyrus. Brain-derived neurotrophic factor mRNA was also measured in the hippocampus by the quantitative polymerase chain reaction. In SHR, PPT had no effect on blood pressure, decreased astrogliosis, slightly increased BDNF mRNA, had no effect on the number of DCX+ progenitors, and increased aromatase staining. Treatment with DPN decreased blood pressure, decreased astrogliosis, increased BDNF mRNA and DCX+ progenitors, and did not modify aromatase staining. We hypothesise that, although both receptor types may participate in the previously reported beneficial effects of 17β-oestradiol in SHR, receptor activation with DPN may preferentially facilitate BDNF mRNA expression and neurogenesis. The results of the present study may help in the design of ER-based neuroprotection for the encephalopathy of hypertension.
Collapse
Affiliation(s)
- L Pietranera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - J Correa
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - M E Brocca
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - P Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - A Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - N Di Giorgio
- Laboratory of Neuroendocrinology, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, Spain
| | - A F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Buenos Aires, Argentina.
- Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
34
|
Effects of ginseng on two main sex steroid hormone receptors: estrogen and androgen receptors. J Ginseng Res 2016; 41:215-221. [PMID: 28413327 PMCID: PMC5386121 DOI: 10.1016/j.jgr.2016.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 07/06/2016] [Accepted: 08/15/2016] [Indexed: 01/01/2023] Open
Abstract
Ginseng has been used in China for at least two millennia and is now popular in over 35 countries. It is one of the world's popular herbs for complementary and alternative medicine and has been shown to have helpful effects on cognition and blood circulation, as well as anti-aging, anti-cancer, and anti-diabetic effects, among many others. The pharmacological activities of ginseng are dependent mainly on ginsenosides. Ginsenosides have a cholesterol-like four trans-ring steroid skeleton with a variety of sugar moieties. Nuclear receptors are one of the most important molecular targets of ginseng, and reports have shown that members of the nuclear receptor superfamily are regulated by a variety of ginsenosides. Here, we review the published literature on the effects of ginseng and its constituents on two main sex steroid hormone receptors: estrogen and androgen receptors. Furthermore, we discuss applications for sex steroid hormone receptor modulation and their therapeutic efficacy.
Collapse
|
35
|
Clarke IJ, Arbabi L. New concepts of the central control of reproduction, integrating influence of stress, metabolic state, and season. Domest Anim Endocrinol 2016; 56 Suppl:S165-79. [PMID: 27345314 DOI: 10.1016/j.domaniend.2016.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 10/21/2022]
Abstract
Gonadotropin releasing hormone is the primary driver of reproductive function and pulsatile GnRH secretion from the brain causes the synthesis and secretion of LH and FSH from the pituitary gland. Recent work has revealed that the secretion of GnRH is controlled at the level of the GnRH secretory terminals in the median eminence. At this level, projections of kisspeptin cells from the arcuate nucleus of the hypothalamus are seen to be closely associated with fibers and terminals of GnRH cells. Direct application of kisspeptin into the median eminence causes release of GnRH. The kisspeptin cells are activated at the time of a natural "pulse" secretion of GnRH, as reflected in the secretion of LH. This appears to be due to input to the kisspeptin cells from glutamatergic cells in the basal hypothalamus, indicating that more than 1 neural element is involved in the secretion of GnRH. Because the GnRH secretory terminals are outside the blood-brain barrier, factors such as kisspeptin may be administered systemically to cause GnRH secretion; this offers opportunities for manipulation of the reproductive axis using factors that do not cross the blood-brain barrier. In particular, kisspeptin or analogs of the same may be used to activate reproduction in the nonbreeding season of domestic animals. Another brain peptide that influences reproductive function is gonadotropin inhibitory hormone (GnIH). Work in sheep shows that this peptide acts on GnRH neuronal perikarya, but projections to the median eminence also allow secretion into the hypophysial portal blood and action of GnIH on pituitary gonadotropes. GnIH cells are upregulated in anestrus, and infusion of GnIH can block the ovulatory surge in GnRH and/or LH secretion. Metabolic status may also affect the secretion of reproduction, and this could involve action of gut peptides and leptin. Neuropeptide Y and Y-receptor ligands have a negative impact on reproduction, and Neuropeptide Y production is markedly increased in negative energy balance; this may be the cause of lowered GnRH and gonadotropin secretion in this state. There is a complex interaction between appetite-regulating peptide neurons and kisspeptin neurons that enables the former to regulate the latter both positively and negatively. In terms of how GnRH secretion is reduced during stress, recent data indicate that GnIH cells are integrally involved, with increased input to the GnRH cells. The secretion of GnIH into the portal blood is not increased during stress, so the negative effect is most likely effected at the level of GnRH neuronal cell bodies.
Collapse
Affiliation(s)
- I J Clarke
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia.
| | - L Arbabi
- Department of Physiology, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
36
|
Lai YJ, Yu D, Zhang JH, Chen GJ. Cooperation of Genomic and Rapid Nongenomic Actions of Estrogens in Synaptic Plasticity. Mol Neurobiol 2016; 54:4113-4126. [PMID: 27324789 PMCID: PMC5509832 DOI: 10.1007/s12035-016-9979-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022]
Abstract
Neuroplasticity refers to the changes in the molecular and cellular processes of neural circuits that occur in response to environmental experiences. Clinical and experimental studies have increasingly shown that estrogens participate in the neuroplasticity involved in cognition, behavior, and memory. It is generally accepted that estrogens exert their effects through genomic actions that occur over a period of hours to days. However, emerging evidence indicates that estrogens also rapidly influence the neural circuitry through nongenomic actions. In this review, we provide an overview of the genomic and nongenomic actions of estrogens and discuss how these actions may cooperate in synaptic plasticity. We then summarize the role of epigenetic modifications, synaptic protein synthesis, and posttranslational modifications, and the splice variants of estrogen receptors in the complicated network of estrogens. The combination of genomic and nongenomic mechanisms endows estrogens with considerable diversity in modulating neural functions including synaptic plasticity.
Collapse
Affiliation(s)
- Yu-Jie Lai
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College of Central South University, Haikou Municipal Hospital, Haikou, Hainan, 570208, China
| | - Dan Yu
- Department of Neurology, Affiliated Haikou Hospital of Xiangya Medical College of Central South University, Haikou Municipal Hospital, Haikou, Hainan, 570208, China
| | - John H Zhang
- Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Guo-Jun Chen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, China.
| |
Collapse
|
37
|
Moore AM, Campbell RE. The neuroendocrine genesis of polycystic ovary syndrome: A role for arcuate nucleus GABA neurons. J Steroid Biochem Mol Biol 2016; 160:106-17. [PMID: 26455490 DOI: 10.1016/j.jsbmb.2015.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/25/2015] [Accepted: 10/02/2015] [Indexed: 12/12/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent and distressing endocrine disorder lacking a clearly identified aetiology. Despite its name, PCOS may result from impaired neuronal circuits in the brain that regulate steroid hormone feedback to the hypothalamo-pituitary-gonadal axis. Ovarian function in all mammals is controlled by the gonadotropin-releasing hormone (GnRH) neurons, a small group of neurons that reside in the pre-optic area of the hypothalamus. GnRH neurons drive the secretion of the gonadotropins from the pituitary gland that subsequently control ovarian function, including the production of gonadal steroid hormones. These hormones, in turn, provide important feedback signals to GnRH neurons via a hormone sensitive neuronal network in the brain. In many women with PCOS this feedback pathway is impaired, resulting in the downstream consequences of the syndrome. This review will explore what is currently known from clinical and animal studies about the identity, relative contribution and significance of the individual neuronal components within the GnRH neuronal network that contribute to the pathophysiology of PCOS. We review evidence for the specific neuronal pathways hypothesised to mediate progesterone negative feedback to GnRH neurons, and discuss the potential mechanisms by which androgens may evoke disruptions in these circuits at different developmental time points. Finally, this review discusses data providing compelling support for disordered progesterone-sensitive GABAergic input to GnRH neurons, originating specifically within the arcuate nucleus in prenatal androgen induced forms of PCOS.
Collapse
Affiliation(s)
- Aleisha M Moore
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand.
| |
Collapse
|
38
|
Parhar IS, Soga T, Ogawa S. Editorial: Reproductive Neuroendocrinology and Social Behavior. Front Neurosci 2016; 10:124. [PMID: 27147941 PMCID: PMC4829586 DOI: 10.3389/fnins.2016.00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 03/14/2016] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ishwar S Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Kuala Lumpur, Malaysia
| | - Tomoko Soga
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Kuala Lumpur, Malaysia
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba Tsukuba, Japan
| |
Collapse
|
39
|
Effects of Prepubertal or Adult Site-Specific Knockdown of Estrogen Receptor β in the Medial Preoptic Area and Medial Amygdala on Social Behaviors in Male Mice. eNeuro 2016; 3:eN-NWR-0155-15. [PMID: 27066533 PMCID: PMC4819287 DOI: 10.1523/eneuro.0155-15.2016] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 11/21/2022] Open
Abstract
Testosterone, after being converted to estradiol in the brain, acts on estrogen receptors (ERα and ERβ) and controls the expression of male-type social behavior. Previous studies in male mice have revealed that ERα expressed in the medial preoptic area (MPOA) and medial amygdala (MeA) are differently involved in the regulation of sexual and aggressive behaviors by testosterone action at the time of testing in adult and/or on brain masculinization process during pubertal period. However, a role played by ERβ in these brain regions still remains unclear. Here we examined the effects of site-specific knockdown of ERβ (βERKD) in the MPOA and MeA on male social behaviors with the use of adeno-associated viral mediated RNA interference methods in ICR/Jcl mice. Prepubertal βERKD in the MPOA revealed that continuous suppression of ERβ gene expression throughout the pubertal period and adulthood decreased aggressive but not sexual behavior tested as adults. Because βERKD in the MPOA only in adulthood did not affect either sexual or aggressive behaviors, it was concluded that pubertal ERβ in the MPOA might have an essential role for the full expression of aggressive behavior in adulthood. On the other hand, although neither prepubertal nor adult βERKD in the MeA had any effects on sexual and aggressive behavior, βERKD in adulthood disrupted sexual preference of receptive females over nonreceptive females. Collectively, these results suggest that ERβ in the MPOA and MeA are involved in the regulation of male sexual and aggressive behavior in a manner substantially different from that of ERα.
Collapse
|
40
|
Bowlby DA, Brown TJ, Hochberg RB, MacLusky NJ. In vitro Autoradiographic Analysis of Regional Changes in Estrogen Receptor Alpha in the Brains of Cycling Female Rats. Neuroendocrinology 2016; 103:538-51. [PMID: 26422138 DOI: 10.1159/000441077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 09/15/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS The contributions of the three principal ovarian steroid hormones (estradiol, progesterone and testosterone) to the regulation of estrogen receptor alpha (ERα) levels in the rat brain were examined during the estrous cycle. METHODS Receptor concentrations were measured using an in vitro autoradiographic technique designed to separately quantify free, unoccupied receptors and receptors 'occupied' by (bound to) endogenous hormone. RESULTS ERα occupation increased at proestrus and declined at estrus, reflecting changes in circulating estradiol and testosterone levels. Total ERα content followed a pattern that was the inverse of the occupation data, falling over the night of proestrus. Between 2.00 and 10.00 a.m. on the day of estrus, total ERα concentrations recovered in all brain regions except the ventromedial nucleus (VMN), in which ERα binding remained depressed at estrus. Administration of the progesterone antagonist mifepristone on the afternoon of proestrus resulted in recovery of ERα levels in the VMN by the morning of estrus, consistent with the hypothesis that the preovulatory progesterone surge selectively inhibits VMN ERα expression. Residual ERα occupation observed at estrus, when estradiol is not detectable in the serum, likely reflects intracranial aromatization of circulating androgens, since the pattern of receptor occupation observed at this stage of the cycle could be reproduced in ovariectomized rats by replacement with testosterone. CONCLUSION These findings indicate that ERα binding in the brain fluctuates during the rat estrous cycle in a region-specific manner and suggest that local aromatization of testosterone may contribute significantly to ERα occupation when circulating estradiol levels are low.
Collapse
Affiliation(s)
- Deborah A Bowlby
- Department of Biomedical Sciences, University of Guelph, Guelph, Ont., Canada
| | | | | | | |
Collapse
|
41
|
Kim HY, Ko KJ, Nam SY, Jeong HJ, Kim HM. The Sound of a Buk (Korean Traditional Drum) Attenuates Anaphylactic Reactions by the Activation of Estrogen Receptor-β. Int Arch Allergy Immunol 2015; 167:242-9. [PMID: 26418257 DOI: 10.1159/000439567] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/20/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Anaphylaxis is associated with systemic vasodilation that causes low blood pressure and induces hypoxic brain damage. The sound of a Buk (Korean traditional drum) is similar to the human heart beat and affects blood pressure, heart rate, and the nervous system by increasing physiological excitation and sympathetic nervous system activity. So, this study focused on the effect of Buk music as a means of treating anaphylaxis. METHODS Mice were given an intraperitoneal injection of compound 48/80 (6.5 mg/kg, a mast cell degranulator). After compound 48/80 injection, mice were exposed to Buk music and white noise for 5 min in a sound isolation booth. The mortality rate was checked over the next 40 min. Levels of hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) in the serum and brain tissues were analyzed by Western blotting, quantitative real-time PCR, and ELISA methods. RESULTS Exposure to Buk music significantly reduced compound 48/80-induced mortality and histamine release, as well as HIF-1α and VEGF levels compared with the compound 48/80 group or compound 48/80 and white noise group. Buk music also reduced levels of tumor necrosis factor-α, and significantly increased estrogen receptor-β mRNA levels. CONCLUSION These results indicate that Buk music has potential for the treatment of anaphylaxis.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
42
|
Abstract
The endocrine hypothalamus constitutes those cells which project to the median eminence and secrete neurohormones into the hypophysial portal blood to act on cells of the anterior pituitary gland. The entire endocrine system is controlled by these peptides. In turn, the hypothalamic neuroendocrine cells are regulated by feedback signals from the endocrine glands and other circulating factors. The neuroendocrine cells are found in specific regions of the hypothalamus and are regulated by afferents from higher brain centers. Integrated function is clearly complex and the networks between and amongst the neuroendocrine cells allows fine control to achieve homeostasis. The entry of hormones and other factors into the brain, either via the cerebrospinal fluid or through fenestrated capillaries (in the basal hypothalamus) is important because it influences the extent to which feedback regulation may be imposed. Recent evidence of the passage of factors from the pars tuberalis and the median eminence casts a new layer in our understanding of neuroendocrine regulation. The function of neuroendocrine cells and the means by which pulsatile secretion is achieved is best understood for the close relationship between gonadotropin releasing hormone and luteinizing hormone, which is reviewed in detail. The secretion of other neurohormones is less rigid, so the relationship between hypothalamic secretion and the relevant pituitary hormones is more complex.
Collapse
Affiliation(s)
- I J Clarke
- Monash University, Department of Physiology, Clayton, Australia
| |
Collapse
|
43
|
Zannoni GF, Ciucci A, Marucci G, Travaglia D, Stigliano E, Foschini MP, Scambia G, Gallo D. Sexual dimorphism in medulloblastoma features. Histopathology 2015; 68:541-8. [DOI: 10.1111/his.12770] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/27/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Gian Franco Zannoni
- Department of Histopathology; Catholic University of the Sacred Heart; Rome Italy
| | - Alessandra Ciucci
- Department of Female, Maternal, Newborn, Child and Adolescent Health; Catholic University of the Sacred Heart; Rome Italy
| | - Gianluca Marucci
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM); Section of Pathology ‘M. Malpighi’; Bellaria Hospital; University of Bologna; Bologna Italy
| | - Daniele Travaglia
- Department of Female, Maternal, Newborn, Child and Adolescent Health; Catholic University of the Sacred Heart; Rome Italy
| | - Egidio Stigliano
- Department of Histopathology; Catholic University of the Sacred Heart; Rome Italy
| | - Maria Pia Foschini
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM); Section of Pathology ‘M. Malpighi’; Bellaria Hospital; University of Bologna; Bologna Italy
| | - Giovanni Scambia
- Department of Female, Maternal, Newborn, Child and Adolescent Health; Catholic University of the Sacred Heart; Rome Italy
| | - Daniela Gallo
- Department of Female, Maternal, Newborn, Child and Adolescent Health; Catholic University of the Sacred Heart; Rome Italy
| |
Collapse
|
44
|
Activation of GPR30 attenuates chronic pain-related anxiety in ovariectomized mice. Psychoneuroendocrinology 2015; 53:94-107. [PMID: 25614360 DOI: 10.1016/j.psyneuen.2014.12.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 12/24/2014] [Accepted: 12/29/2014] [Indexed: 11/21/2022]
Abstract
Estrogen regulates neuroendocrine and inflammatory processes that play critical roles in neuroinflammation, anxiety, and chronic pain. Patients suffering from chronic pain often complain of anxiety. However, limited information is available regarding the neural circuitry of chronic pain-related anxiety and the related function of estrogen. Hindpaw injection of complete Freund's adjuvant (CFA) and chronic constriction injury (CCI) of the sciatic nerve induced notable pain sensitization and anxiety-like behavior in ovariectomized (OVX) mice. We found that the level of G-protein-coupled receptor 30 (GPR30), a membrane estrogen receptor, was significantly increased in the basolateral amygdala (BLA) of ovariectomized (OVX) mice suffering from chronic inflammatory and neuropathic pain. Subcutaneous injection or BLA local infusion of the GPR30 agonist G1 significantly reduced anxiety-like behavior in CFA-injected and CCI-OVX mice; however, this treatment did not alter the nociceptive threshold. GPR30 knock down by shRNA in the BLA of OVX mice inhibited the anxiolytic effects of GPR30 activation. G1 administration reversed the upregulation of GluR1 subunit in AMPA and NR2A-containing NMDA receptors and the downregulation of GABAA receptors in the BLA of CFA-injected and CCI-OVX mice. Electrophysiological recording revealed that GPR30 activation could prevent imbalance between excitatory and inhibitory transmissions in the BLA synapses of CFA-injected OVX mice. In conclusion, GPR30 activation induced anxiolytic effects but did not affect the nociceptive threshold of mice under chronic pain. The anxiolytic effects of GPR30 were partially due to maintaining the balance between excitatory and inhibitory transmissions in the BLA.
Collapse
|
45
|
Litwa E, Rzemieniec J, Wnuk A, Lason W, Krzeptowski W, Kajta M. Apoptotic and neurotoxic actions of 4-para-nonylphenol are accompanied by activation of retinoid X receptor and impairment of classical estrogen receptor signaling. J Steroid Biochem Mol Biol 2014; 144 Pt B:334-47. [PMID: 25092517 DOI: 10.1016/j.jsbmb.2014.07.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 11/30/2022]
Abstract
4-para-Nonylphenol (NP) is a non-ionic surfactant that has widespread and uncontrolled distribution in the environment. Little is known, however, about its actions on neuronal cells during critical developmental periods. This study aimed to investigate the mechanisms underlying the apoptotic and toxic actions of NP on mouse embryonic neuronal cells and the possible interactions of NP with estrogen receptor (ER)- and retinoid X receptor (RXR)-mediated intracellular signaling. Treatment of mouse hippocampal neuronal cell cultures with NP (5 and 10μM) induced apoptotic and neurotoxic effects. The 2 and 7 day-old mouse hippocampal cultures were vulnerable to 5 and 10μM NP, whereas 12 day-old cultures responded only to the highest concentration of NP, thus suggesting an age-dependent action of the chemical on neuronal cells. The use of specific inhibitors did not support the involvement of calpains in NP-induced apoptosis, but indicated caspase-8- and caspase-9-dependent effects of NP. Specific ER antagonists MPP and PHTPP potentiated the NP-induced loss of mitochondrial membrane potential and increase in lactate dehydrogenase (LDH) release whereas, ER agonists PPT and DPN inhibited these effects. RXR antagonist HX531 diminished the NP-evoked loss of mitochondrial membrane potential, the activity of caspase-3 and LDH release. In addition, exposure to NP inhibited ERα- and ERβ-specific immunofluorescence but stimulated RXR-specific immunolabeling in mouse hippocampal cells. In conclusion, our study demonstrated that the apoptotic and toxic actions of NP on neuronal cells in early development is accompanied by an impairment of ER- and stimulation of RXR-mediated signaling pathways. Taking into account NP-induced alterations in mRNA expression levels of particular types of RXRs, we suggest that NP affected mainly RXRα and RXRβ, but not RXRγ signaling.
Collapse
Affiliation(s)
- E Litwa
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - J Rzemieniec
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - A Wnuk
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - W Lason
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland
| | - W Krzeptowski
- Department of Cell Biology and Imaging, Confocal Microscopy Laboratory, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland
| | - M Kajta
- Department of Experimental Neuroendocrinology, Institute of Pharmacology, Polish Academy of Sciences, 12 Smetna Street, 31-343 Krakow, Poland.
| |
Collapse
|
46
|
Tsuda MC, Yamaguchi N, Nakata M, Ogawa S. Modification of female and male social behaviors in estrogen receptor beta knockout mice by neonatal maternal separation. Front Neurosci 2014; 8:274. [PMID: 25228857 PMCID: PMC4151037 DOI: 10.3389/fnins.2014.00274] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 08/14/2014] [Indexed: 01/20/2023] Open
Abstract
Maternal separation (MS) is an animal model mimicking the effects of early life stress on the development of emotional and social behaviors. Recent studies revealed that MS stress increased social anxiety levels in female mice and reduced peri-pubertal aggression in male mice. Estrogen receptor (ER) β plays a pivotal role in the regulation of stress responses and anxiety-related and social behaviors. Behavioral studies using ERβ knockout (βERKO) mice reported increased social investigation and decreased social anxiety in βERKO females, and elevated aggression levels in βERKO males compared to wild-type (WT) mice. In the present study, using βERKO and WT mice, we examined whether ERβ contributes to MS effects on anxiety and social behaviors. βERKO and WT mice were separated from their dam daily (4 h) from postnatal day 1–14 and control groups were left undisturbed. First, MS and ERβ gene deletion individually increased anxiety-related behaviors in the open field test, but only in female mice. Anxiety levels were not further modified in βERKO female mice subjected to MS stress. Second, βERKO female mice showed higher levels of social investigation compared with WT in the social investigation test and long-term social preference test. However, MS greatly reduced social investigation duration and elevated number of stretched approaches in WT and βERKO females in the social investigation test, suggesting elevated levels of social anxiety in both genotypes. Third, peri-pubertal and adult βERKO male mice were more aggressive than WT mice as indicated by heightened aggression duration. On the other hand, MS significantly decreased aggression duration in both genotypes, but only in peri-pubertal male mice. Altogether, these results suggest that βERKO mice are sensitive to the adverse effects of MS stress on subsequent female and male social behaviors, which could then have overrode the ERβ effects on female social anxiety and male aggression.
Collapse
Affiliation(s)
- Mumeko C Tsuda
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba Tsukuba, Japan
| | - Naoko Yamaguchi
- Department of Pharmacology, School of Medicine, Aichi Medical University Nagakute, Japan
| | - Mariko Nakata
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba Tsukuba, Japan
| | - Sonoko Ogawa
- Laboratory of Behavioral Neuroendocrinology, University of Tsukuba Tsukuba, Japan
| |
Collapse
|
47
|
Nguyen CT, Luong TT, Kim GL, Pyo S, Rhee DK. Korean Red Ginseng inhibits apoptosis in neuroblastoma cells via estrogen receptor β-mediated phosphatidylinositol-3 kinase/Akt signaling. J Ginseng Res 2014; 39:69-75. [PMID: 25535479 PMCID: PMC4268566 DOI: 10.1016/j.jgr.2014.06.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 06/02/2014] [Accepted: 06/12/2014] [Indexed: 02/07/2023] Open
Abstract
Background Ginseng has been shown to exert antistress effects both in vitro and in vivo. However, the effects of ginseng on stress in brain cells are not well understood. This study investigated how Korean Red Ginseng (KRG) controls hydrogen peroxide-induced apoptosis via regulation of phosphatidylinositol-3 kinase (PI3K)/Akt and estrogen receptor (ER)-β signaling. Methods Human neuroblastoma SK-N-SH cells were pretreated with KRG and subsequently exposed to H2O2. The ability of KRG to inhibit oxidative stress-induced apoptosis was assessed in MTT cytotoxicity assays. Apoptotic protein expression was examined by Western blot analysis. The roles of ER-β, PI3K, and p-Akt signaling in KRG regulation of apoptosis were studied using small interfering RNAs and/or target antagonists. Results Pretreating SK-N-SH cells with KRG decreased expression of the proapoptotic proteins p-p53 and caspase-3, but increased expression of the antiapoptotic protein BCL2. KRG pretreatment was also associated with increased ER-β, PI3K, and p-Akt expression. Conversely, ER-β inhibition with small interfering RNA or inhibitor treatment increased p-p53 and caspase-3 levels, but decreased BCL2, PI3K, and p-Akt expression. Moreover, inhibition of PI3K/Akt signaling diminished p-p53 and caspase-3 levels, but increased BCL2 expression. Conclusion Collectively, the data indicate that KRG represses oxidative stress-induced apoptosis by enhancing PI3K/Akt signaling via upregulation of ER-β expression.
Collapse
Affiliation(s)
| | | | - Gyu-Lee Kim
- School of Pharmacy, Sungkyunkwan University, Su-Won, Korea
| | - Suhkneung Pyo
- School of Pharmacy, Sungkyunkwan University, Su-Won, Korea
| | - Dong-Kwon Rhee
- School of Pharmacy, Sungkyunkwan University, Su-Won, Korea
| |
Collapse
|
48
|
Clarke IJ. Interface between metabolic balance and reproduction in ruminants: focus on the hypothalamus and pituitary. Horm Behav 2014; 66:15-40. [PMID: 24568750 DOI: 10.1016/j.yhbeh.2014.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 02/13/2014] [Accepted: 02/15/2014] [Indexed: 01/24/2023]
Abstract
This article is part of a Special Issue "Energy Balance". The interface between metabolic regulators and the reproductive system is reviewed with special reference to the sheep. Even though sheep are ruminants with particular metabolic characteristics, there is a broad consensus across species in the way that the reproductive system is influenced by metabolic state. An update on the neuroendocrinology of reproduction indicates the need to account for the way that kisspeptin provides major drive to gonadotropin releasing hormone (GnRH) neurons and also mediates the feedback effects of gonadal steroids. The way that kisspeptin function is influenced by appetite regulating peptides (ARP) is considered. Another newly recognised factor is gonadotropin inhibitory hormone (GnIH), which has a dual function in that it suppresses reproductive function whilst also acting as an orexigen. Our understanding of the regulation of food intake and energy expenditure has expanded exponentially in the last 3 decades and historical perspective is provided. The function of the regulatory factors and the hypothalamic cellular systems involved is reviewed with special reference to the sheep. Less is known of these systems in the cow, especially the dairy cow, in which a major fertility issue has emerged in parallel with selection for increased milk production. Other endocrine systems--the hypothalamo-pituitary-adrenal axis, the growth hormone (GH) axis and the thyroid hormones--are influenced by metabolic state and are relevant to the interface between metabolic function and reproduction. Special consideration is given to issues such as season and lactation, where the relationship between metabolic hormones and reproductive function is altered.
Collapse
Affiliation(s)
- Iain J Clarke
- Monash University, Department of Physiology, Wellington Road, Clayton 3168, Australia.
| |
Collapse
|
49
|
Cheong RY, Porteous R, Chambon P, Abrahám I, Herbison AE. Effects of neuron-specific estrogen receptor (ER) α and ERβ deletion on the acute estrogen negative feedback mechanism in adult female mice. Endocrinology 2014; 155:1418-27. [PMID: 24476134 DOI: 10.1210/en.2013-1943] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The negative feedback mechanism through which 17β-estradiol (E2) acts to suppress the activity of the GnRH neurons remains unclear. Using inducible and cell-specific genetic mouse models, we examined the estrogen receptor (ER) isoforms expressed by neurons that mediate acute estrogen negative feedback. Adult female mutant mice in which ERα was deleted from all neurons in the neonatal period failed to exhibit estrous cycles or negative feedback. Adult mutant female mice with neonatal neuronal ERβ deletion exhibited normal estrous cycles, but a failure of E2 to suppress LH secretion was seen in ovariectomized mice. Mutant mice with a GnRH neuron-selective deletion of ERβ exhibited normal cycles and negative feedback, suggesting no critical role for ERβ in GnRH neurons in acute negative feedback. To examine the adult roles of neurons expressing ERα, an inducible tamoxifen-based Cre-LoxP approach was used to ablate ERα from neurons that express calmodulin kinase IIα in adults. This resulted in mice with no estrous cycles, a normal increase in LH after ovariectomy, but an inability of E2 to suppress LH secretion. Finally, acute administration of ERα- and ERβ-selective agonists to adult ovariectomized wild-type mice revealed that activation of ERα suppressed LH secretion, whereas ERβ agonists had no effect. This study highlights the differences in adult reproductive phenotypes that result from neonatal vs adult ablation of ERα in the brain. Together, these experiments expand previous global knockout studies by demonstrating that neurons expressing ERα are essential and probably sufficient for the acute estrogen negative feedback mechanism in female mice.
Collapse
Affiliation(s)
- Rachel Y Cheong
- Centre for Neuroendocrinology (R.Y.C., R.P., I.A., A.E.H.), Department of Physiology, University of Otago School of Medical Sciences, Dunedin 9054, New Zealand; and Institut de Génétique et de Biologie Moléculaire et Cellulaire (P.C.), 67400 Illkirch, France
| | | | | | | | | |
Collapse
|
50
|
Handa RJ, Weiser MJ. Gonadal steroid hormones and the hypothalamo-pituitary-adrenal axis. Front Neuroendocrinol 2014; 35:197-220. [PMID: 24246855 PMCID: PMC5802971 DOI: 10.1016/j.yfrne.2013.11.001] [Citation(s) in RCA: 299] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 10/04/2013] [Accepted: 11/07/2013] [Indexed: 12/17/2022]
Abstract
The hypothalamo-pituitary-adrenal (HPA) axis represents a complex neuroendocrine feedback loop controlling the secretion of adrenal glucocorticoid hormones. Central to its function is the paraventricular nucleus of the hypothalamus (PVN) where neurons expressing corticotropin releasing factor reside. These HPA motor neurons are a primary site of integration leading to graded endocrine responses to physical and psychological stressors. An important regulatory factor that must be considered, prior to generating an appropriate response is the animal's reproductive status. Thus, PVN neurons express androgen and estrogen receptors and receive input from sites that also express these receptors. Consequently, changes in reproduction and gonadal steroid levels modulate the stress response and this underlies sex differences in HPA axis function. This review examines the make up of the HPA axis and hypothalamo-pituitary-gonadal (HPG) axis and the interactions between the two that should be considered when exploring normal and pathological responses to environmental stressors.
Collapse
Affiliation(s)
- Robert J Handa
- Department of Basic Medical Science, The University of Arizona College of Medicine, Phoenix, AZ 85004, United States.
| | - Michael J Weiser
- DSM Nutritional Products Ltd., R&D Human Nutrition and Health, Boulder, CO 80301, United States
| |
Collapse
|