1
|
Trout AL, McLouth CJ, Westberry JM, Sengoku T, Wilson ME. Estrogen's sex-specific effects on ischemic cell death and estrogen receptor mRNA expression in rat cortical organotypic explants. AGING BRAIN 2024; 5:100117. [PMID: 38650743 PMCID: PMC11033203 DOI: 10.1016/j.nbas.2024.100117] [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: 10/19/2023] [Revised: 02/14/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024] Open
Abstract
Estrogens, such as the biologically active 17-β estradiol (E2), regulate not only reproductive behaviors in adults, but also influence neurodevelopment and neuroprotection in both females and males. E2, contingent upon the timing and concentration of the therapy, is neuroprotective in female and male rodent models of stroke. In Vivo studies suggest that E2 may partially mediate this neuroprotection, particularly in the cortex, via ERα. In Vitro studies, utilizing a chemically induced ischemic injury in cortical explants from both sexes, suggest that ERα or ERβ signaling is needed to mediate the E2 protection. Since we know that the timing and concentration of E2 therapy may be sex-specific, we examined if E2 (1 nM) mediates neuroprotection when female and male cortical explants are separately isolated from postnatal day (PND) 3-4 rat. Changes in basal levels ERα, ERβ, and AR mRNA expression are compared across early post-natal development in the intact cortex and the corresponding days in vitro (DIV) for cortical explants. Following ischemic injury at 7 DIV, cell death and ERα, ERβ and AR mRNA expression was compared in female and male cortical explants. We provide evidence that E2-mediated protection is maintained in isolated cortical explants from females, but not male rats. In female cortical explants, the E2-mediated protection at 24 h occurs secondarily to a blunted transient increase in ERα mRNA at 12 h. These results suggest that cortical E2-mediated protection is influenced by sex and supports data to differentially treat females and males following ischemic injury.
Collapse
Affiliation(s)
- Amanda L. Trout
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher J McLouth
- Department of Neurology, University of Kentucky, Lexington, KY, 40536, USA
- Department of Biostatistics, University of Kentucky, Lexington, KY, 40536, USA
| | - Jenne M. Westberry
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Tomoko Sengoku
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Melinda E. Wilson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
2
|
Li LH, Ling DD, Lin H, Wang ZC, Sun ZR, Zhang YQ, Yang L, Zhang J, Cao H. Ovariectomy induces hyperalgesia accompanied by upregulated estrogen receptor α and protein kinase B in the rat spinal cord. Physiol Behav 2023; 271:114342. [PMID: 37673233 DOI: 10.1016/j.physbeh.2023.114342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/20/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Hormone supplementation is one of the common therapies for menopause-related disorders. Among different tools, the ovariectomy (OVX) rodents are widely accepted as an appropriate menopausal pain model. Our previous study has showed that OVX produces a significant pain facilitation in both acute pain and tonic pain, however, the underlying mechanisms remain unclear. In this study, we examined the effects of OVX treatment and estradiol (E2) supplementation on formalin-induced nociceptive responses, and explored the associated spinal mechanisms. Female Sprague-Dawley rats underwent bilateral OVX, and E2 supplementation was given subcutaneously from the 5th week after surgery (30 μg/day for 7 days). Our results showed that formalin-induced nociceptive behaviors did not differ between diestrus and proestrus stages of the estrous in intact rats. However, OVX exacerbated formalin-evoked inflammatory pain, especially in the late phase at 4-5 weeks but not 2 weeks post-surgery. E2 supplementation significantly reversed the OVX-triggered hyperalgesia. Double immunofluorescence staining revealed that both ERα and ERβ in the spinal dorsal horn were co-labeled with the neuronal markers, but not with markers of astrocytes or microglia. The spinal ERα (but not ERβ) expression significantly increased in the OVX group, which was reversed by E2 supplementation. Moreover, the OVX individuals showed an increased protein kinase B (AKT) level in lumbar spinal cord, and E2 supplementation diminished the AKT expression in OVX rats. Finally, intrathecal injection Wortmannin, an inhibitor for AKT signaling, effectively reduced the nociceptive behaviors in the late phase and the number of c-fos positive cells. Together, our findings indicate that E2 supplementation alleviates the OVX-induced hyperalgesia, which might be involved in spinal ERα and AKT mechanisms.
Collapse
Affiliation(s)
- Li-Hong Li
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Dan-Dan Ling
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Hong Lin
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Zhe-Chen Wang
- Department of Psychology, School of Social Development and Public Policy, Fudan University, Shanghai 200032, China
| | - Zhi-Rong Sun
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China
| | - Yu-Qiu Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Liu Yang
- Shanghai Dunlu Biomedical Technology Co. Ltd. Shanghai 201611, China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, Shanghai, China.
| | - Hong Cao
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| |
Collapse
|
3
|
Johnson CS, Mermelstein PG. The interaction of membrane estradiol receptors and metabotropic glutamate receptors in adaptive and maladaptive estradiol-mediated motivated behaviors in females. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:33-91. [PMID: 36868633 DOI: 10.1016/bs.irn.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Estrogen receptors were initially identified as intracellular, ligand-regulated transcription factors that result in genomic change upon ligand binding. However, rapid estrogen receptor signaling initiated outside of the nucleus was also known to occur via mechanisms that were less clear. Recent studies indicate that these traditional receptors, estrogen receptor α and estrogen receptor β, can also be trafficked to act at the surface membrane. Signaling cascades from these membrane-bound estrogen receptors (mERs) can rapidly alter cellular excitability and gene expression, particularly through the phosphorylation of CREB. A principal mechanism of neuronal mER action has been shown to occur through glutamate-independent transactivation of metabotropic glutamate receptors (mGlu), which elicits multiple signaling outcomes. The interaction of mERs with mGlu has been shown to be important in many diverse functions in females, including driving motivated behaviors. Experimental evidence suggests that a large part of estradiol-induced neuroplasticity and motivated behaviors, both adaptive and maladaptive, occurs through estradiol-dependent mER activation of mGlu. Herein we will review signaling through estrogen receptors, both "classical" nuclear receptors and membrane-bound receptors, as well as estradiol signaling through mGlu. We will focus on how the interactions of these receptors and their downstream signaling cascades are involved in driving motivated behaviors in females, discussing a representative adaptive motivated behavior (reproduction) and maladaptive motivated behavior (addiction).
Collapse
Affiliation(s)
- Caroline S Johnson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Paul G Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
4
|
Immunohistochemical Detection of Estrogen Receptor-Beta (ERβ) with PPZ0506 Antibody in Murine Tissue: From Pitfalls to Optimization. Biomedicines 2022; 10:biomedicines10123100. [PMID: 36551855 PMCID: PMC9775465 DOI: 10.3390/biomedicines10123100] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022] Open
Abstract
The estrogen receptor beta (ERβ) is physiologically essential for reproductive biology and is implicated in various diseases. However, despite more than 20 years of intensive research on ERβ, there are still uncertainties about its distribution in tissues and cellular expression. Several studies show contrasts between mRNA and protein levels, and the use of knockout strategies revealed that many commercially available antibodies gave false-positive expression results. Recently, a specific monoclonal antibody against human ERβ (PPZ0506) showed cross-reactivity with rodents and was optimized for the detection of rat ERβ. Herein, we established an immunohistochemical detection protocol for ERβ protein in mouse tissue. Staining was optimized on murine ovaries, as granulosa cells are known to strongly express ERβ. The staining results were confirmed by western blot analysis and RT-PCR. To obtain accurate and reliable staining results, different staining conditions were tested in paraffin-embedded tissues. Different pitfalls were encountered in immunohistochemical detection. Strong heat-induced epitope retrieval (HIER) and appropriate antibody dilution were required to visualize specific nuclear expression of ERβ. Finally, the specificity of the antibody was confirmed by using ovaries from Esr2-depleted mice. However, in some animals, strong (non-specific) background staining appeared. These signals could not be significantly alleviated with commercially available additional blocking solutions and are most likely due to estrus-dependent expression of endogenous immunoglobulins. In summary, our study showed that the antibody PPZ0506, originally directed against human ERβ, is also suitable for reliable detection of murine ERβ. An established staining protocol mitigated ambiguities regarding the expression and distribution of ERβ in different tissues and will contribute to an improved understanding of its role and functions in murine tissues in the future.
Collapse
|
5
|
Sex Differences in Psychostimulant Abuse: Implications for Estrogen Receptors and Histone Deacetylases. Genes (Basel) 2022; 13:genes13050892. [PMID: 35627277 PMCID: PMC9140379 DOI: 10.3390/genes13050892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Substance abuse is a chronic pathological disorder that negatively affects many health and neurological processes. A growing body of literature has revealed gender differences in substance use. Compared to men, women display distinct drug-use phenotypes accompanied by recovery and rehabilitation disparities. These observations have led to the notion that sex-dependent susceptibilities exist along the progression to addiction. Within this scope, neuroadaptations following psychostimulant exposure are thought to be distinct for each sex. This review summarizes clinical findings and animal research reporting sex differences in the subjective and behavioral responses to cocaine, methamphetamine, and nicotine. This discussion is followed by an examination of epigenetic and molecular alterations implicated in the addiction process. Special consideration is given to histone deacetylases and estrogen receptor-mediated gene expression.
Collapse
|
6
|
Zhang M, Weiland H, Schöfbänker M, Zhang W. Estrogen Receptors Alpha and Beta Mediate Synaptic Transmission in the PFC and Hippocampus of Mice. Int J Mol Sci 2021; 22:ijms22031485. [PMID: 33540803 PMCID: PMC7867372 DOI: 10.3390/ijms22031485] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
Distinct from ovarian estradiol, the steroid hormone 17ß-estradiol (E2) is produced in the brain and is involved in numerous functions, particularly acting as a neurosteroid. However, the physiological role of E2 and the mechanism of its effects are not well known. In hippocampal slices, 17ß-estradiol has been found to cause a modest increase in fast glutamatergic transmission; because some of these effects are rapid and acute, they might be mediated by membrane-associated receptors via nongenomic action. Moreover, activation of membrane estrogen receptors can rapidly modulate neuron function in a sex-specific manner. To further investigate the neurological role of E2, we examined the effect of E2, as an estrogen receptor (ER) agonist, on synaptic transmission in slices of the prefrontal cortex (PFC) and hippocampus in both male and female mice. Whole-cell recordings of spontaneous excitatory postsynaptic currents (sEPSC) in the PFC showed that E2 acts as a neuromodulator in glutamatergic transmission in the PFC in both sexes, but often in a cell-specific manner. The sEPSC amplitude and/or frequency responded to E2 in three ways, namely by significantly increasing, decreasing or having no response. Additional experiments using an agonist selective for ERß, diarylpropionitrile (DPN) showed that in males the sEPSC and spontaneous inhibitory postsynaptic currents sIPSC responses were similar to their E2 responses, but in females the estrogen receptor ß (ERß) agonist DPN did not influence excitatory transmission in the PFC. In contrast, in the hippocampus of both sexes E2 potentiated the gluatmatergic synaptic transmission in a subset of hippocampal cells. These data indicate that activation of E2 targeting probably a estrogen subtypes or different downstream signaling affect synaptic transmission in the brain PFC and hippocampus between males versus females mice.
Collapse
Affiliation(s)
- Mingyue Zhang
- Correspondence: ; Tel.: +49-2518-351-824; Fax: +49-2518-357-123
| | | | | | | |
Collapse
|
7
|
Hughes CHK, Murphy BD. Nuclear receptors: Key regulators of somatic cell functions in the ovulatory process. Mol Aspects Med 2020; 78:100937. [PMID: 33288229 DOI: 10.1016/j.mam.2020.100937] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 12/30/2022]
Abstract
The development of the ovarian follicle to its culmination by ovulation is an essential element of fertility. The final stages of ovarian follicular growth are characterized by granulosa cell proliferation and differentiation, and steroid synthesis under the influence of follicle-stimulating hormone (FSH). The result is a population of granulosa cells poised to respond to the ovulatory surge of luteinizing hormone (LH). Members of the nuclear receptor superfamily of transcription factors play indispensable roles in the regulation of these events. The key regulators of the final stages of follicular growth that precede ovulation from this family include the estrogen receptor beta (ESR2) and the androgen receptor (AR), with additional roles for others, including steroidogenic factor-1 (SF-1) and liver receptor homolog-1 (LRH-1). Following the LH surge, the mural and cumulus granulosa cells undergo rapid changes that result in expansion of the cumulus layer, and a shift in ovarian steroid hormone biosynthesis from estradiol to progesterone production. The nuclear receptor best associated with these events is LRH-1. Inadequate cumulus expansion is also observed in the absence of AR and ESR2, but not the progesterone receptor (PGR). The terminal stages of ovulation are regulated by PGR, which increases the abundance of the proteases that are directly responsible for rupture. It further regulates the prostaglandins and cytokines associated with the inflammatory-like characteristics of ovulation. LRH-1 regulates PGR, and is also a key regulator of steroidogenesis, cellular proliferation, and cellular migration, and cytoskeletal remodeling. In summary, nuclear receptors are among the panoply of transcriptional regulators with roles in ovulation, and several are necessary for normal ovarian function.
Collapse
Affiliation(s)
- Camilla H K Hughes
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Qc, J2S 2M2, Canada
| | - Bruce D Murphy
- Centre de Recherche en Reproduction et Fertilité, Université de Montréal, St-Hyacinthe, Qc, J2S 2M2, Canada.
| |
Collapse
|
8
|
Involvement of the MEN1 Gene in Hormone-Related Cancers: Clues from Molecular Studies, Mouse Models, and Patient Investigations. ENDOCRINES 2020. [DOI: 10.3390/endocrines1020007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MEN1 mutation predisposes patients to multiple endocrine neoplasia type 1 (MEN1), a genetic syndrome associated with the predominant co-occurrence of endocrine tumors. Intriguingly, recent evidence has suggested that MEN1 could also be involved in the development of breast and prostate cancers, two major hormone-related cancers. The first clues as to its possible role arose from the identification of the physical and functional interactions between the menin protein, encoded by MEN1, and estrogen receptor α and androgen receptor. In parallel, our team observed that aged heterozygous Men1 mutant mice developed cancerous lesions in mammary glands of female and in the prostate of male mutant mice at low frequencies, in addition to endocrine tumors. Finally, observations made both in MEN1 patients and in sporadic breast and prostate cancers further confirmed the role played by menin in these two cancers. In this review, we present the currently available data concerning the complex and multifaceted involvement of MEN1 in these two types of hormone-dependent cancers.
Collapse
|
9
|
Yare K, Woodward M. Hormone Therapy and Effects on Sporadic Alzheimer’s Disease in Postmenopausal Women: Importance of Nomenclature. J Alzheimers Dis 2020; 73:23-37. [DOI: 10.3233/jad-190896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Katrine Yare
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| | - Michael Woodward
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| |
Collapse
|
10
|
Arao Y, Korach KS. Transactivation Function-1-Mediated Partial Agonist Activity of Selective Estrogen Receptor Modulator Requires Homo-Dimerization of the Estrogen Receptor α Ligand Binding Domain. Int J Mol Sci 2019; 20:ijms20153718. [PMID: 31366023 PMCID: PMC6695978 DOI: 10.3390/ijms20153718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 11/16/2022] Open
Abstract
The isolation of estrogen receptor alpha (ERα) cDNA was successful around 30 years ago. The characteristics of ERα protein have been examined from various aspects, primarily through in vitro cell culture studies, but more recently using in vivo experimental models. There remains, however, some uncharacterized ERα functionalities. In particular, the mechanism of partial agonist activity of selective estrogen receptor modulators (SERMs) that involves control of the N-terminal transcription function of ERα, termed AF-1, is still an unsolved ERα functionality. We review the possible mechanism of SERM-dependent regulation of ERα AF-1-mediated transcriptional activity, which includes the role of helix 12 of ERα ligand binding domain (LBD) for SERM-dependent AF-1 regulation. In addition, we describe a specific portion of the LBD that associates with blocking AF-1 activity with an additional role of the F-domain in mediating SERM activity.
Collapse
Affiliation(s)
- Yukitomo Arao
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Durham, NC 27709, USA.
| | - Kenneth S Korach
- Receptor Biology Section, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences/NIH, Durham, NC 27709, USA.
| |
Collapse
|
11
|
Micevych PE, Sinchak K. Extranuclear signaling by ovarian steroids in the regulation of sexual receptivity. Horm Behav 2018; 104:4-14. [PMID: 29753716 PMCID: PMC6240501 DOI: 10.1016/j.yhbeh.2018.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/02/2018] [Accepted: 05/03/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Paul E Micevych
- Dept of Neurobiology, David Geffen School of Medicine at UCLA, Laboratory of Neuroendocrinology of the UCLA Brain Research Institute, United States
| | - Kevin Sinchak
- Dept of Biological Sciences, California State University, Long Beach, United States.
| |
Collapse
|
12
|
Sèdes L, Thirouard L, Maqdasy S, Garcia M, Caira F, Lobaccaro JMA, Beaudoin C, Volle DH. Cholesterol: A Gatekeeper of Male Fertility? Front Endocrinol (Lausanne) 2018; 9:369. [PMID: 30072948 PMCID: PMC6060264 DOI: 10.3389/fendo.2018.00369] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/19/2018] [Indexed: 12/14/2022] Open
Abstract
Cholesterol is essential for mammalian cell functions and integrity. It is an important structural component maintaining the permeability and fluidity of the cell membrane. The balance between synthesis and catabolism of cholesterol should be tightly regulated to ensure normal cellular processes. Male reproductive function has been demonstrated to be dependent on cholesterol homeostasis. Here we review data highlighting the impacts of cholesterol homeostasis on male fertility and the molecular mechanisms implicated through the signaling pathways of some nuclear receptors.
Collapse
|
13
|
Shahin S, Singh SP, Chaturvedi CM. Mobile phone (1800MHz) radiation impairs female reproduction in mice, Mus musculus, through stress induced inhibition of ovarian and uterine activity. Reprod Toxicol 2017; 73:41-60. [PMID: 28780396 DOI: 10.1016/j.reprotox.2017.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/27/2017] [Accepted: 08/01/2017] [Indexed: 12/30/2022]
Abstract
Present study investigated the long-term effects of mobile phone (1800MHz) radiation in stand-by, dialing and receiving modes on the female reproductive function (ovarian and uterine histo-architecture, and steroidogenesis) and stress responses (oxidative and nitrosative stress). We observed that mobile phone radiation induces significant elevation in ROS, NO, lipid peroxidation, total carbonyl content and serum corticosterone coupled with significant decrease in antioxidant enzymes in hypothalamus, ovary and uterus of mice. Compared to control group, exposed mice exhibited reduced number of developing and mature follicles as well as corpus lutea. Significantly decreased serum levels of pituitary gonadotrophins (LH, FSH), sex steroids (E2 and P4) and expression of SF-1, StAR, P-450scc, 3β-HSD, 17β-HSD, cytochrome P-450 aromatase, ER-α and ER-β were observed in all the exposed groups of mice, compared to control. These findings suggest that mobile phone radiation induces oxidative and nitrosative stress, which affects the reproductive performance of female mice.
Collapse
Affiliation(s)
- Saba Shahin
- Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Surya Pal Singh
- Department of Electronics Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi 221005, India
| | | |
Collapse
|
14
|
Liu KC, Lau SW, Ge W. Spatiotemporal expression analysis of nuclear estrogen receptors in the zebrafish ovary and their regulation in vitro by endocrine hormones and paracrine factors. Gen Comp Endocrinol 2017; 246:218-225. [PMID: 28013034 DOI: 10.1016/j.ygcen.2016.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/11/2016] [Accepted: 12/19/2016] [Indexed: 11/20/2022]
Abstract
Estradiol (E2) stimulates luteinizing hormone receptor (lhcgr) expression via nuclear estrogen receptors (nERs) in the zebrafish ovary. We have demonstrated that endocrine hormones such as gonadotropin (hCG) and paracrine factors such as epidermal growth factor (EGF) and pituitary adenylate cyclase-activating peptide (PACAP) can modulate E2-induced lhcgr expression in vitro. These observations raised a question on whether these hormones and factors exert their effects via regulating the expression of nERs. In this study, we first characterized the spatiotemporal expression profiles of three nER subtypes in the zebrafish ovary, including esr1 (ERα), esr2a (ERβ2) and esr2b (ERβ1). All three nERs increased their expression at the pre-vitellogenic stage and peaked at mid- (esr1 and esr2a) or late vitellogenic (esr2b) stage, followed by a significant decline at the full-grown stage. RT-PCR analysis showed that esr1 and esr2b were exclusively expressed in the follicle layer while esr2a was expressed in both compartments. We then examined how E2, hCG, PACAP and EGF regulated the expression of nERs in cultured zebrafish follicle cells. E2 quickly increased esr1 but reduced esr2a and esr2b expression from 1.5 to 12h of treatment. Similarly, EGF down-regulated esr2a significantly at 1.5h and this effect was further intensified at 24h. hCG decreased the expression of all three nER subtypes with similar potency throughout the 24-h time-course. Interestingly, PACAP exerted a biphasic regulation on esr2a. Our present study suggests that nERs, especially esr2a, provide potential target points for other hormones and factors to modulate E2 activity during folliculogenesis in the zebrafish.
Collapse
Affiliation(s)
- Ka-Cheuk Liu
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Shuk-Wa Lau
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wei Ge
- Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, China; School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| |
Collapse
|
15
|
Mowa CN, Papka RE. The Role of Sensory Neurons in Cervical Ripening: Effects of Estrogen and Neuropeptides. J Histochem Cytochem 2016; 52:1249-58. [PMID: 15385571 DOI: 10.1177/002215540405201001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Central nervous system nuclei and circuits, such as the medial preoptic, ventromedial and paraventricular nuclei of the hypothalamus, play important roles in reproduction and parturition, and are influenced by estrogen. Peripheral autonomic and sensory neurons also play important roles in pregnancy and parturition. Moreover, the steroid hormone estrogen acts directly, not only on the reproductive tract organs (uterus and cervix), but also on the central and peripheral nerves by regulating expression of various neuronal genes. The peripheral primary afferent neurons innervating the uterine cervix relay mechanical and biochemical sensory information induced by local cervical events and by passage of fetuses, to the spinal cord and supraspinal centers. Consequently, the birth process in mammals is influenced by the combined action of neurons and hormones. Peripheral sensory stimuli, induced physiologically by fetal expulsion or mechanically by vaginocervical stimulation, alter behavior, as well as autonomic and neuroendocrine systems. Recent evidence indicates that primary afferent neurons innervating the cervix, in addition to their sensory effects, likely exert local “efferent” actions on the ripening cervix near term. These efferent effects may involve estrogen-regulated production of such neuropeptides as substance P and calcitonin gene-related peptide in lumbosacral dorsal root ganglia, and their release in the cervix. Collectively, these findings suggest an interrelationship among estrogen, cervix-related sensory neurons, and local cervical events near term.
Collapse
Affiliation(s)
- C N Mowa
- Northeastern Ohio Universities College of Medicine, Department of Neurobiology, 4209 State Rt. 44, P.O. Box 95, Rootstown, OH 44272, USA
| | | |
Collapse
|
16
|
Sołtysik K, Czekaj P. ERα36--Another piece of the estrogen puzzle. Eur J Cell Biol 2015; 94:611-25. [PMID: 26522827 DOI: 10.1016/j.ejcb.2015.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 09/03/2015] [Accepted: 10/09/2015] [Indexed: 12/22/2022] Open
Abstract
Although the nuclear action of estrogen receptors (ER) is a well-known fact, evidence supporting membrane estrogen receptors is steadily accumulating. New ER variants of unrecognized function have been discovered. ERα is a product of the ESR1 gene. It serves not only as a template for the full-length 66kDa protein, but also for smaller isoforms which exist as independent receptors. The recently discovered ERα36 (36kDa), consisting of 310 amino acids of total 595 ERα66 protein residues, is an example of that group. The transcription initiation site is identified in the first intron of the ESR1 gene. C-Terminal 27 amino acids are encoded by previously unknown exon 9. The presence of this unique C-terminal sequence creates an opportunity for the production of selective antibodies. ERα36 has been shown to have a high affinity to the cell membrane and as much as 90% of the protein can be bound with it. Post-translational palmitoylation is suspected to play a crucial role in ERα36 anchoring to the cell membrane. In silico analysis suggests the existence of a potential transmembrane domain in ERα36. ERα36 was found in most cells of animals at various ages, but its exact physiological function remains to be fully elucidated. It seems that cells traditionally considered as being deprived of ER are able to respond to hormonal stimulation via the ERα36 receptor. Moreover, ERα36 displays unique pharmacological properties and its action may be behind antiestrogen resistance. The use of ERα36 in cancer diagnosis gives rise to great expectations.
Collapse
Affiliation(s)
- Kamil Sołtysik
- Students Scientific Society, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Piotr Czekaj
- Department of Cytophysiology, Chair of Histology and Embryology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland.
| |
Collapse
|
17
|
Characterization of the fundamental properties of the N-terminal truncation (Δ exon 1) variant of estrogen receptor α in the rat. Gene 2015; 571:117-25. [DOI: 10.1016/j.gene.2015.06.086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/02/2015] [Accepted: 06/22/2015] [Indexed: 02/05/2023]
|
18
|
Le Grand A, Bouter A, Couturier A, Mulner-Lorillon O, Le Goff X, Chesnel F, Sire O, Le Tilly V. Investigation of the functional properties and subcellular localization of alpha human and rainbow trout estrogen receptors within a unique yeast cellular context. J Steroid Biochem Mol Biol 2015; 149:17-26. [PMID: 25595040 DOI: 10.1016/j.jsbmb.2015.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/26/2014] [Accepted: 01/11/2015] [Indexed: 12/22/2022]
Abstract
Estrogens are steroid hormones that play a pivotal role in growth, differentiation and function of reproductive and non-reproductive tissues, mediated through estrogen receptors (ERs). Estrogens are involved in different genomic and non-genomic cell signaling pathways which involve well-defined subcellular ER localizations. Thus, ER activity results from complex interplays between intrinsic binding properties and specific subcellular localization. Since these two factors are deeply intricate, we carried out, in a unique yeast cell context, a comparative study to better understand structure/function/subcellular distribution relationships. This was carried out by comparing two ERs: the human ER α subtype (hERα) and the short form of the α isoform of the rainbow trout ER (rtERαS). Their distinct binding properties to agonist and antagonist ligands and subcellular localizations were characterized in Saccharomyces cerevisiae yeast cells. An unexpected partial agonistic effect of ICI 182-780 was observed for rtERαS. Concomitant to distinct binding properties, distinct subcellular localizations were observed before and after ligand stimulation. Due to the unique cell context, the link between ERs intrinsic binding properties and subcellular localizations is partly unveiled and issues are hypothesized based on the role of cytoplasmic transient complexes which play a role in the ER cytoplasmic/nuclear partition, which in turn is critical for the recruitment of co-regulators in the nucleus.
Collapse
Affiliation(s)
- Adélaïde Le Grand
- Laboratoire d'Ingénierie des Matériaux de Bretagne, Université de Bretagne-Sud, CER Yves Coppens, BP573, 56017 Vannes Cedex, France
| | - Anthony Bouter
- Molecular Imaging and NanoBioTechnology, UMR 5248 CBMN, CNRS-Université Bordeaux 1-ENITAB, IECB, 2 rue Robert Escarpit, 33607 Pessac, France
| | - Anne Couturier
- CNRS/Université de Rennes 1, Institut de Génétique & Développement de Rennes, UMR 6290, 2 Ave. Prof. Léon Bernard, CS 34317, 35043 Rennes Cedex, France
| | - Odile Mulner-Lorillon
- CNRS/UPMC Université Paris 06, UMR 8227 LBI2M, Traduction, Cycle Cellulaire et Développement, Station Biologique, CS 90074, 29688 Roscoff Cedex, France
| | - Xavier Le Goff
- CNRS/Université de Rennes 1, Institut de Génétique & Développement de Rennes, UMR 6290, 2 Ave. Prof. Léon Bernard, CS 34317, 35043 Rennes Cedex, France
| | - Franck Chesnel
- CNRS/Université de Rennes 1, Institut de Génétique & Développement de Rennes, UMR 6290, 2 Ave. Prof. Léon Bernard, CS 34317, 35043 Rennes Cedex, France
| | - Olivier Sire
- Laboratoire d'Ingénierie des Matériaux de Bretagne, Université de Bretagne-Sud, CER Yves Coppens, BP573, 56017 Vannes Cedex, France
| | - Véronique Le Tilly
- Laboratoire d'Ingénierie des Matériaux de Bretagne, Université de Bretagne-Sud, CER Yves Coppens, BP573, 56017 Vannes Cedex, France.
| |
Collapse
|
19
|
Contrò V, R. Basile J, Proia P. Sex steroid hormone receptors, their ligands, and nuclear and non-nuclear pathways. AIMS MOLECULAR SCIENCE 2015. [DOI: 10.3934/molsci.2015.3.294] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
20
|
Rouiller-Fabre V, Guerquin MJ, N’Tumba-Byn T, Muczynski V, Moison D, Tourpin S, Messiaen S, Habert R, Livera G. Nuclear receptors and endocrine disruptors in fetal and neonatal testes: a gapped landscape. Front Endocrinol (Lausanne) 2015; 6:58. [PMID: 25999913 PMCID: PMC4423451 DOI: 10.3389/fendo.2015.00058] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/07/2015] [Indexed: 11/28/2022] Open
Abstract
During the last decades, many studies reported that male reproductive disorders are increasing among humans. It is currently acknowledged that these abnormalities can result from fetal exposure to environmental chemicals that are progressively becoming more concentrated and widespread in our environment. Among the chemicals present in the environment (air, water, food, and many consumer products), several can act as endocrine disrupting compounds (EDCs), thus interfering with the endocrine system. Phthalates, bisphenol A (BPA), and diethylstilbestrol (DES) have been largely incriminated, particularly during the fetal and neonatal period, due to their estrogenic and/or anti-androgenic properties. Indeed, many epidemiological and experimental studies have highlighted their deleterious impact on fetal and neonatal testis development. As EDCs can affect many different genomic and non-genomic pathways, the mechanisms underlying the adverse effects of EDC exposure are difficult to elucidate. Using literature data and results from our laboratory, in the present review, we discuss the role of classical nuclear receptors (genomic pathway) in the fetal and neonatal testis response to EDC exposure, particularly to phthalates, BPA, and DES. Among the nuclear receptors, we focused on some of the most likely candidates, such as peroxisome-proliferator activated receptor (PPAR), androgen receptor (AR), estrogen receptors (ERα and β), liver X receptors (LXR), and small heterodimer partner (SHP). First, we describe the expression and potential functions (based on data from studies using receptor agonists and mouse knockout models) of these nuclear receptors in the developing testis. Then, for each EDC studied, we summarize the main evidences indicating that the reprotoxic effect of each EDC under study is mediated through a specific nuclear receptor(s). We also point-out the involvement of other receptors and nuclear receptor-independent pathways.
Collapse
Affiliation(s)
- Virginie Rouiller-Fabre
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
- *Correspondence: Virginie Rouiller-Fabre, Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, CEA, DSV, iRCM, SCSR, LDG, BP6, Fontenay aux Roses F-92265, France,
| | - Marie Justine Guerquin
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Thierry N’Tumba-Byn
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Vincent Muczynski
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Delphine Moison
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Sophie Tourpin
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Sébastien Messiaen
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - René Habert
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| | - Gabriel Livera
- Unit of Genetic Stability, Stem Cells and Radiation, Laboratory of Development of the Gonads, Sorbonne Paris Cité, Université Paris Diderot, Fontenay-aux-Roses, France
- CEA, DSV, iRCM, SCSR, LDG, Fontenay-aux-Roses, France
- Unité 967, INSERM, Fontenay aux Roses, France
| |
Collapse
|
21
|
Liu H, Wang L, Ma H, Guo R, Kang R, Han J, Dong Z. Coumestrol inhibits carotid sinus baroreceptor activity by cAMP/PKA dependent nitric oxide release in anesthetized male rats. Biochem Pharmacol 2014; 93:42-8. [PMID: 25449602 DOI: 10.1016/j.bcp.2014.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/02/2014] [Accepted: 11/05/2014] [Indexed: 12/16/2022]
Abstract
Phytoestrogens could offer multiple beneficial effects on the cardiovascular system. Here, we have examined the effects of coumestrol (CMT) on carotid baroreceptors activity (CBA) and the possible mechanisms in male rats. The functional parameters of carotid baroreceptors were measured by recording sinus nerve afferent discharge in anesthetized male rats with perfused isolated carotid sinus. The levels of protein expression were determined by using ELISA and Western blotting. CMT (1 to 100μmolL(-1)) inhibited CBA, which shifted the functional curve of the carotid baroreceptor to the right and downward, with a marked decrease in the peak slope and the peak integral value of carotid sinus nerve discharge in a concentration dependent manner. These effects were not blocked by a specific estrogen receptor antagonist ICI 182,780, but were completely abolished by nitric oxide (NO) synthase inhibitor l-NAME (N(G)-nitro-l-arginine methyl ester). Furthermore, a NO donor, SIN-1(3-morpholion-sydnon-imine), could potentiate these inhibitory effects of CMT. CMT stimulated the phosphorylation of Ser(1176)-eNOS (endothelial nitric oxide synthase) in a dose-dependent manner in carotid bifurcation tissue over a perfusion period of 15min. The rapid activation of eNOS by CMT was blocked by a highly selective PKA (protein kinase A) inhibitor H89. In addition, inhibition of PI3K (phosphatidylinositol-3-kinase) and ERK (extracellular signal-regulated kinase) pathways had no effect on eNOS activation by CMT. CMT inhibited CBA via eNOS activation and NO synthesis. These effects were mediated by the cAMP/PKA pathway and were unrelated to the estrogenic effect.
Collapse
Affiliation(s)
- Haitao Liu
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lixuan Wang
- Department of Histology and Embryology, Hebei Medical University, Shijiazhuang, China
| | - Huijuan Ma
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Rong Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Rongtian Kang
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianmin Han
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenming Dong
- Department of Anesthesiology, the Second Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
22
|
Urano T, Inoue S. Genetics of osteoporosis. Biochem Biophys Res Commun 2014; 452:287-93. [DOI: 10.1016/j.bbrc.2014.07.141] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 07/18/2014] [Indexed: 01/22/2023]
|
23
|
Lee HS, Sasagawa SI, Kato S, Fukuda R, Horiuchi H, Ohta A. Yeast Two-Hybrid Detection Systems That Are Highly Sensitive to a Certain Kind of Endocrine Disruptors. Biosci Biotechnol Biochem 2014; 70:521-4. [PMID: 16495672 DOI: 10.1271/bbb.70.521] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We tested the effects of several combinations of bait and fish components of the yeast two-hybrid detection system for estrogenic activity. A combination of the full-length human estrogen receptor alpha with the nuclear receptor-binding domain of co-activator steroid receptor co-activator-1 (SRC-1) or transcriptional intermediate factor-2 (TIF-2) was most effective for estrogen-dependent induction of the chromosome-integrated UAS(GAL)-CYC1(p)-lacZ reporter construct among the two-hybrid systems so far tested.
Collapse
|
24
|
Li W, Zhang J, Mu W, Wen H. Cloning, characterization and expression of estrogen receptor beta in the male half-smooth tongue sole, Cynoglossus semilaevis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2013; 39:671-682. [PMID: 23053612 DOI: 10.1007/s10695-012-9729-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 09/25/2012] [Indexed: 06/01/2023]
Abstract
A full-length sequence encoding the estrogen receptor beta was isolated from half-smooth tongue sole, Cynoglossus semilaevis (hstsERβ) using reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends procedures. The hstsERβ cDNA clone was found to contain 1,791 nucleotides including an open reading frame that encodes 578 amino acids. The deduced hstsERβ protein consisted of six nuclear receptor-characteristic domains. Based on a phylogenetic analysis, the hstsERβ C and E domains are highly conserved compared to other fishes. The potential phosphorylation sites for PKC, CK-2 and PTK are also found in this protein. Highest amino acid identities were found for hstsERβ with common carp (Cyprinus carpio) ERβ (76 %) and Japanese flounder (Paralichthys olivaceus) ERβ (76 %). Tissue expression analysis confirmed that the hstsERβ was widely distributed and predominantly expressed in testis, brain and liver. Seasonal changes in the testis, brain and liver expression profiles of hstsERβ were examined by RT-PCR; the present results suggest that level of hstsERβ in brain increased to the highest then decreases with gonadal growth; whereas in the testis and liver, the hstsERβ mRNA level dropped to lowest then slightly increased.
Collapse
Affiliation(s)
- Wenge Li
- Fisheries College, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | | | | | | |
Collapse
|
25
|
Wilson ME, Sengoku T. Developmental regulation of neuronal genes by DNA methylation: environmental influences. Int J Dev Neurosci 2013; 31:448-51. [PMID: 23501000 DOI: 10.1016/j.ijdevneu.2013.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 03/04/2013] [Accepted: 03/06/2013] [Indexed: 12/27/2022] Open
Abstract
Steroid hormones have wide-ranging organizational, activational and protective actions in the brain. In particular, the organizational effects of early exposure to 17β-estradiol (E2) and glucocorticoids are essential for long-lasting behavioral and cognitive functions. Both steroid hormones mediate many of their actions through intracellular receptors that act as transcription factors. In the rodent cerebral cortex, estrogen receptor mRNA and protein expression are high early in postnatal life and declines dramatically as the animal approaches puberty. An understanding of the molecular mechanisms driving this developmental regulation of gene expression is critical for understanding the complex events that determine lasting brain physiology and prime the plasticity of neurons. Gene expression can be suppressed by the epigenetic modification of the promoter regions by DNA methylation that results in gene silencing. Indeed, the decrease in ERα mRNA expression in the cortex during development is accompanied by an increase in promoter methylation. Numerous environmental stimuli can alter the DNA methylation that occurs for ERα, glucocorticoid receptors, as well as many other critical genes involved in neuronal development. For example, maternal behavior toward pups can alter epigenetic regulation of ERα mRNA expression. Additionally perinatal stress and exposure to environmental estrogens can also have lasting effects on gene expression by modifying DNA methylation of these important genes. Taken together, there appears to be a critical window during development where, outside factors that alter epigenetic programming can have lasting effects on neuronal gene expression.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, United States.
| | | |
Collapse
|
26
|
Hiroi H, Momoeda M, Watanabe T, Ito M, Ikeda K, Tsutsumi R, Hosokawa Y, Koizumi M, Zenri F, Muramatsu M, Taketani Y, Inoue S. Expression and regulation of transient receptor potential cation channel, subfamily M, member 2 (TRPM2) in human endometrium. Mol Cell Endocrinol 2013; 365:146-52. [PMID: 23142700 DOI: 10.1016/j.mce.2012.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 09/09/2012] [Accepted: 10/12/2012] [Indexed: 12/27/2022]
Abstract
To identify estrogen-responsive genes, we previously isolated estrogen receptor (ER)-binding DNA fragments from human genomic DNA using a recombinant ER protein. Six DNA fragments, each including a perfect palindromic estrogen response element (ERE), were obtained. The nucleotide sequence of one of the six fragments (E1 fragment) showed that the ERE of the E1 fragment is located in the 3'-untranslated region (UTR) of transient receptor potential cation channel, subfamily M, member 2 (TRPM2). Here, we confirmed the estrogen-dependent enhancer activity of the ERE of the E1 fragment by chloramphenicol acetyltransferase assay. TRPM2 mRNA expression was investigated in human endometrium, cultured human endometrial stromal cells (ESCs), and cultured human endometrial epithelial cells (EECs) using RT-PCR. Quantitative RT-PCR revealed that TRPM2 mRNA expression in ESCs increased after 17β-estradiol (E2) treatment. This study demonstrated for the first time that TRPM2 is an estrogen-responsive gene expressed in human endometrial cells.
Collapse
Affiliation(s)
- Hisahiko Hiroi
- Department of Obstetrics and Gynecology, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Ishii H, Kobayashi M, Munetomo A, Miyamoto T, Sakuma Y. Novel splicing events and post-transcriptional regulation of human estrogen receptor α E isoforms. J Steroid Biochem Mol Biol 2013; 133:120-8. [PMID: 23032375 DOI: 10.1016/j.jsbmb.2012.09.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 09/21/2012] [Accepted: 09/22/2012] [Indexed: 12/01/2022]
Abstract
Expression of the estrogen receptor α (ERα) gene is subject to complex regulation. To elucidate the mechanisms of this regulation, the genomic organization and the physiological role of the multiple 5'-untranslated regions (5'-UTRs) must be determined. Here, we investigated the expression and splicing patterns of the human ERα E isoforms. We identified two novel untranslated exons, N1 and N2, in the 5'-region of the human ERα gene and multiple E isoform mRNA variants generated by alternative usage of non-coding internal exons. Expression of the N1-containing variants was observed only in the human breast adenocarcinoma cell line, MCF7, while the N2-containing variants were expressed in the adult liver and MCF7 cells. We examined post-transcriptional regulation of the variant mRNAs using luciferase reporter assays and quantitative PCR. The insertion of untranslated internal exons into the 5'-UTRs of the E isoforms reduced their translation efficiency, but barely influenced mRNA turnover. Our results indicate that the genomic organization of the human ERα gene and the splicing profiles of the human ERα E isoforms are more complicated than previously reported. Furthermore, the 5'-UTRs of the E isoforms post-transcriptionally control human ERα expression mainly through translational repression.
Collapse
Affiliation(s)
- Hirotaka Ishii
- Department of Physiology, Nippon Medical School, 1 Sendagi, Tokyo 113-8602, Japan.
| | | | | | | | | |
Collapse
|
28
|
McClure RES, Barha CK, Galea LAM. 17β-Estradiol, but not estrone, increases the survival and activation of new neurons in the hippocampus in response to spatial memory in adult female rats. Horm Behav 2013; 63:144-57. [PMID: 23063473 DOI: 10.1016/j.yhbeh.2012.09.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/18/2012] [Accepted: 09/22/2012] [Indexed: 10/27/2022]
Abstract
Estrogens fluctuate across the lifespan in women, with circulating 17β-estradiol levels higher pre-menopause than estrone and circulating estrone levels higher postmenopause than 17β-estradiol. Estrone is a common component of hormone replacement therapies, but research shows that 17β-estradiol may have a greater positive impact on cognition. Previous studies show that acute estrone and 17β-estradiol impact hippocampus-dependent learning and cell proliferation in the dentate gyrus in a dose-dependent manner in adult female rats. The current study explores how chronic treatment with estrone and 17β-estradiol differentially influences spatial learning, hippocampal neurogenesis and activation of new neurons in response to spatial memory. Adult female rats received daily injections of vehicle (sesame oil), or a 10 μg dose of either 17β-estradiol or estrone for 20 days. One day following the first hormone injection all rats were injected with the DNA synthesis marker, bromodeoxyuridine. On days 11-15 after BrdU injection rats were trained on a spatial reference version of the Morris water maze, and five days later (day 20 of estrogens treatment) were given a probe trial to assess memory retention. Cell proliferation was assessed by the endogenous cell cycle marker, Ki67, cell survival was assessed by counting the number and density of BrdU-ir cells in the dentate gyrus and cell activation was assessed by the percentage of BrdU-ir cells that were co-labelled with the immediate early gene product zif268. There were no significant differences between groups in acquisition or retention of Morris water maze. However, the 17β-estradiol group had significantly higher, while the estrone group had significantly lower, levels of cell survival (BrdU-ir cells) in the dentate gyrus compared to controls. Furthermore, rats injected with 17β-estradiol showed significantly higher levels of activation of new neurons in response to spatial memory compared to controls. These results provide insight into how estrogens differentially influence the brain and behavior, and may provide insight into the development of hormone replacement therapies for women.
Collapse
Affiliation(s)
- Robyn E S McClure
- Department of Psychology, Graduate Program in Neuroscience, Brain Research Centre, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|
29
|
Nugent BM, Tobet SA, Lara HE, Lucion AB, Wilson ME, Recabarren SE, Paredes AH. Hormonal programming across the lifespan. Horm Metab Res 2012; 44:577-86. [PMID: 22700441 PMCID: PMC3756611 DOI: 10.1055/s-0032-1312593] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Hormones influence countless biological processes across an animal's lifespan. Many hormone-mediated events occur within developmental sensitive periods, during which hormones have the potential to cause permanent tissue-specific alterations in anatomy and physiology. There are numerous selective critical periods in development with different targets being affected during different periods. This review outlines the proceedings of the Hormonal Programming in Development session at the US-South American Workshop in Neuroendocrinology in August 2011. Here we discuss how gonadal steroid hormones impact various biological processes within the brain and gonads during early development and describe the changes that take place in the aging female ovary. At the cellular level, hormonal targets in the brain include neurons, glia, or vasculature. On a genomic/epigenomic level, transcription factor signaling and epigenetic changes alter the expression of critical hormone receptor genes across development and following ischemic brain insult. In addition, organizational hormone exposure alters epigenetic processes in specific brain nuclei and may be an important mediator of sexual differentiation of the neonatal brain. Brain targets of hormonal programming, such as the paraventricular nucleus of the hypothalamus, may be critical in influencing the development of peripheral targets, such as the ovary. Exposure to excess hormones can cause abnormalities in the ovary during development leading to polycystic ovarian syndrome (PCOS). Exposure to excess androgens during fetal development also has a profound effect on the development of the male reproductive system. In addition, increased activity of the sympathetic nerve and stress during early life have been linked to PCOS symptomology in adulthood. Finally, we describe how age-related decreases in fertility are linked to high levels of nerve growth factor (NGF), which enhances sympathetic nerve activity and alters ovarian function.
Collapse
Affiliation(s)
- B M Nugent
- University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Cao DY, Ji Y, Tang B, Traub RJ. Estrogen receptor β activation is antinociceptive in a model of visceral pain in the rat. THE JOURNAL OF PAIN 2012; 13:685-94. [PMID: 22698981 DOI: 10.1016/j.jpain.2012.04.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/19/2012] [Accepted: 04/27/2012] [Indexed: 01/31/2023]
Abstract
UNLABELLED The mechanism underlying estrogen modulation of visceral pain remains unclear. Our previous studies indicate that activation of estrogen receptor α (ERα) enhances visceral pain. The purpose of the present study was to investigate the role of estrogen receptor β (ERβ) activation in spinal processing of visceral stimuli. The effects of selective ERβ agonists on the visceromotor response (VMR) and dorsal horn neuronal responses to colorectal distention (CRD) were tested in ovariectomized and intact female rats. The magnitude of the VMR to CRD was significantly attenuated by ERβ agonists diarylpropionitrile (DPN) and WAY-200070 4 hours after subcutaneous injection. Pretreatment with the estrogen receptor antagonist ICI 182,780 obscured the DPN-evoked attenuation. There was no effect of DPN on the VMR at earlier time points. Subcutaneous and spinal administration of DPN attenuated the response of visceroceptive dorsal horn neurons with a comparable time course. DPN attenuated the VMR in intact rats regardless of estrous cycle stage. The time course of effect of ERβ activation on the visceromotor response and neuronal activity is consistent with transcriptional or translational modulation of neuronal activity. PERSPECTIVE Activation of ERβ is antinociceptive in the colorectal distention model of visceral pain, which may provide a therapeutic target to manage irritable bowel syndrome in the clinic.
Collapse
Affiliation(s)
- Dong-Yuan Cao
- Department of Neural and Pain Sciences, School of Dentistry, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | |
Collapse
|
31
|
Paisley JC, Huddleston GG, Carruth LL, Petrulis A, Grober MS, Clancy AN. Sexual responses of the male rat medial preoptic area and medial amygdala to estrogen I: site specific suppression of estrogen receptor alpha. Horm Behav 2012; 62:50-7. [PMID: 22565217 DOI: 10.1016/j.yhbeh.2012.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 03/31/2012] [Accepted: 04/28/2012] [Indexed: 11/17/2022]
Abstract
Male rat copulation is mediated by estrogen-sensitive neurons in the medial preoptic area (MPO) and medial amygdala (MEA); however, the mechanisms through which estradiol (E(2)) acts are not fully understood. We hypothesized that E(2) acts through estrogen receptor α (ERα) in the MPO and MEA to promote male mating behavior. Antisense oligodeoxynucleotides (AS-ODN) complementary to ERα mRNA were bilaterally infused via minipumps into either brain area to block the synthesis of ERα, which we predicted would reduce mating. Western blot analysis and immunocytochemistry revealed a knockdown of ERα expression in each brain region; however, compared to saline controls, males receiving AS-ODN to the MPO showed significant reductions in all components of mating, whereas males receiving AS-ODN to the MEA continued to mate normally. These results suggest that E(2) acts differently in these brain regions to promote the expression of male rat sexual behavior and that ERα in the MPO, but not in the MEA, promotes mating.
Collapse
Affiliation(s)
- Jacquelyn C Paisley
- Department of Biology, Georgia State University, Atlanta, GA 30302‐4010, USA
| | | | | | | | | | | |
Collapse
|
32
|
Mombelli E. Evaluation of the OECD (Q)SAR Application Toolbox for the profiling of estrogen receptor binding affinities. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2012; 23:37-57. [PMID: 22014213 DOI: 10.1080/1062936x.2011.623325] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The determination of binding affinities for the estrogen receptor (ER) is used extensively to assess potential hazards to human health and the environment arising from chemicals that can interfere with natural hormone homeostasis. Given the great number of chemicals to which humans and wildlife are exposed, (quantitative) structure-activity relationship (Q)SAR models for the characterization of ER disruptors represent a fast and cost-efficient alternative to experimental testing. In this toxicological context, the freely available Organisation for Economic Co-operation and Development (OECD) (Q)SAR Application Toolbox provides a profiler for the categorical profiling of chemicals according to their ER binding propensities. The aim of this study was to evaluate the predictive performances of this profiler. To achieve such a purpose, prediction results with the ER-profiler were compared with experimental binding affinities relative to two large datasets of chemicals (rat and human). The resulting Cooper statistics indicated that the binding affinities of the majority of chemicals included in the retained datasets could be correctly predicted.
Collapse
Affiliation(s)
- E Mombelli
- a Unité Modèles pour l'Ecotoxicologie et la Toxicologie (METO), Institut National de l'Environnement Industriel et des Risques (INERIS) , Verneuil-en-Halatte , France
| |
Collapse
|
33
|
Abstract
During aging, there is an increase in neurodegenerative diseases and a decrease in cognitive performance. Postmenopausal women are more vulnerable as their estrogen levels decline, but most hormone replacement therapies do not prevent cognitive decline. One potential reason is that the timing of hormone replacement is critical and changes in the estrogen receptor expression may over-ride hormonal intervention. In rodents, estrogen receptor β (ERβ) mRNA decreases in the cortex with age. One mechanism by which ERβ mRNA could be regulated is by epigenetic modification of ERβ promoter. Here, we show an increase in methylation of ERβ promoter corresponding to decrease in ERβ mRNA in the cortex of an aging female.
Collapse
|
34
|
Lu L, Zhang H, Lv N, Ma X, Tian L, Hu X, Liu S, Xu M, Weng Q, Watanabe G, Taya K. Immunolocalization of Androgen Receptor, Aromatase Cytochrome P450, Estrogen Receptor Alpha and Estrogen Receptor Beta Proteins during the Breeding Season in Scent Glands of Muskrats (Ondatra zibethicus). Zoolog Sci 2011; 28:727-32. [DOI: 10.2108/zsj.28.727] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
35
|
Katsu Y, Lange A, Miyagawa S, Urushitani H, Tatarazako N, Kawashima Y, Tyler CR, Iguchi T. Cloning, expression and functional characterization of carp,Cyprinus carpio, estrogen receptors and their differential activations by estrogens. J Appl Toxicol 2011; 33:41-9. [DOI: 10.1002/jat.1707] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 05/07/2011] [Accepted: 05/07/2011] [Indexed: 12/23/2022]
Affiliation(s)
| | - Anke Lange
- School of Biosciences, College of Life and Environmental Sciences; University of Exeter; Exeter; EX4 4PS; UK
| | | | | | - Norishisa Tatarazako
- National Institute for Environmental Studies; 16-2 Onogawa; Tsukuba; Ibaraki; 305-8506; Japan
| | - Yukio Kawashima
- Japan NUS Co. Ltd, 7-5-25 Nishi-Shinjyuku; Shinjyuku-ku; Tokyo; 160-0023; Japan
| | - Charles R. Tyler
- School of Biosciences, College of Life and Environmental Sciences; University of Exeter; Exeter; EX4 4PS; UK
| | | |
Collapse
|
36
|
Ishii H, Sakuma Y. Complex organization of the 5'-untranslated region of the mouse estrogen receptor α gene: identification of numerous mRNA transcripts with distinct 5'-ends. J Steroid Biochem Mol Biol 2011; 125:211-8. [PMID: 21397022 DOI: 10.1016/j.jsbmb.2011.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 02/18/2011] [Accepted: 03/04/2011] [Indexed: 01/24/2023]
Abstract
The 5'-untranslated region (5'-UTR) of the estrogen receptor α (ERα) gene plays an important role in determining its tissue-specific expression. We examined the 5'-UTRs of the mouse ERα mRNA variants in depth using the Basic Local Alignment Search Tool (BLAST), rapid amplification of 5'-cDNA ends (5'-RACE) and RT-PCR. We demonstrated the presence of multiple variants containing unique 5'-UTRs. We mapped the cDNA sequences onto the mouse genome, and found that both alternative splicing from four different leader exons (A, C, F1, and H) to exon 1, and combinations of 12 internal exons (X1, X2, X3, X4, F2/X5, X6, X7, X8, X9, X10, X11, and B) generate multiple ERα transcripts. Mouse exon B, that has homologies with human exon B and rat exon 0T, was used as an internal exon, not as a leader exon. RT-PCR analysis revealed distinct expression patterns of the variants, suggesting that the alternative promoter usage and alternative splicing are regulated in a tissue-specific manner. Our results indicate that the genomic organization of the mouse ERα gene is complicated as previously shown in the rat ERα gene. In addition, both alternative promoter usage and alternative splicing contribute to the remarkable mRNA diversity of the mouse ERα gene.
Collapse
Affiliation(s)
- Hirotaka Ishii
- Department of Physiology, Nippon Medical School, 1 Sendagi, Bunkyo, Tokyo 113-8602, Japan.
| | | |
Collapse
|
37
|
Jesmin S, Mowa CN, Sultana SN, Mia S, Islam R, Zaedi S, Sakuma I, Hattori Y, Hiroe M, Yamaguchi N. Estrogen receptor alpha and beta are both involved in the cerebral VEGF/Akt/NO pathway and cerebral angiogenesis in female mice. ACTA ACUST UNITED AC 2011; 31:337-46. [PMID: 21187644 DOI: 10.2220/biomedres.31.337] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We have previously demonstrated that vascular endothelial growth factor (VEGF) is critical for cerebral angiogenesis in middle-aged female rats and may play a role in the flow-preserving neuroprotective actions of estrogen through its angiogenic and antiapoptotic properties. Here, we attempt to elucidate the effects of estrogen and the specific estrogen receptor (ER) subtype in cerebral VEGF/Akt/NO pathways and cerebral angiogenesis using 15-week old female mice that are either wild-type (WT), lack estrogen receptor α (ERαKO) or β (ERβKO). Protein levels of VEGF and basic signaling molecules of VEGF angiogenic pathway in the frontal cortex were expressed as follows, as revealed by ELISA and immunoblotting : a) VEGF; WT: ERαKO: ERβKO, 47 ± 15: 27 ± 5: 28 ± 5 pg/mg, respectively (P < 0.01); b) KDR decreased about 40% in both ERαKO and ERβKO compared to WT; c) Akt was significantly down-regulated in both ERαKO and ERβKO compared to WT; d) phosphorylated Akt (pAkt); WT: ERαKO: ERβKO, 0.6 ± 0.2: 0.3 ± 0.01: 0.3 ± 0.1 units/mg, respectively; e) phosphorylated eNOS significantly decreased about 45% in both ERαKO and ERβKO compared to WT. Cerebral capillary density decreased in both ERαKO and ERβKO compared to WT. Thus, it can be concluded that in female mice, VEGF/Akt/eNOS pathway plays an important role in cerebral angiogenesis and that both ER subtypes are involved in the regulation of VEGF and its signaling molecule expression in the frontal cortex.
Collapse
Affiliation(s)
- Subrina Jesmin
- Division of Gene Therapeutics, Research Institute, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim JG, Kim HR, Park YJ, Chung KH, Oh SM. Molecular Cloning and Characterization of the Estrogen Receptor from the Striped Bitterling (Acheilognathus yamatsutae). ENVIRONMENTAL HEALTH AND TOXICOLOGY 2011; 26:e2011005. [PMID: 22125766 PMCID: PMC3214976 DOI: 10.5620/eht.2011.26.e2011005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Accepted: 02/25/2011] [Indexed: 05/31/2023]
Abstract
OBJECTIVES In order to identify the possibility of striped bitterling (SB) (Acheilognathus yamatsutae) being used as a test species for estrogenic endocrine disrupting chemicals (EEDCs), we carried out the cloning and sequence characterization of the estrogen receptor (ER). METHODS The ER from a striped bitterling was obtained by reverse transcriptase-polymerase chain reaction (RT-PCR), 5'- and 3'-rapid amplification of cDNA ends (5'-RACE and 3'-RACE) and T-vector cloning. The expression of ER mRNA was also analyzed in six tissues (brain, liver, kidney, gill, gonad, and intestines) by real-time PCR. RESULTS We obtained an ER from the striped bitterling. The SB ER cDNA was 2189 base pairs (bp) in length and contained a 1707 bp open reading frame that encoded 568 amino acid residues. The SB ER amino acid sequence clustered in a monophyletic group with the ERα of other fish, and was more closely related to zebrafish ERα (88% identity) than to the ERα of other fish. The SB ER cDNA was divided into A/B, C, D, E and F domains. The SB ER has conserved important sequences for ER functions, such as the DNA binding domain (D domain), which are consistent with those of other teleosts. CONCLUSIONS The ER of the striped bitterling could provide basic information in toxicological studies of EEDCs in the striped bitterling.
Collapse
Affiliation(s)
- Jong Geuk Kim
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Ha Ryong Kim
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | - Yong Joo Park
- College of Pharmacy, Sungkyunkwan University, Suwon, Korea
| | | | - Seung Min Oh
- Hoseo Toxicological Research Center, Hoseo University, Asan, Korea
| |
Collapse
|
39
|
Nugent BM, McCarthy MM. Epigenetic underpinnings of developmental sex differences in the brain. Neuroendocrinology 2011; 93:150-8. [PMID: 21411982 PMCID: PMC7068790 DOI: 10.1159/000325264] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 01/28/2011] [Indexed: 12/12/2022]
Abstract
Sexual differentiation of the brain is a crucial developmental process that enables the lifelong expression of sexually dimorphic behaviors, including those necessary for successful reproduction. During a perinatal sensitive period, gonadal hormones defeminize and masculinize the male brain, and a lack of gonadal steroids allows for feminization in the female. This hormonally-induced differentiation permanently alters neural structures, creating highly dimorphic brain regions; however, the mechanism by which hormones exert their long-lasting effects are still largely unknown. Epigenetic processes such as DNA methylation and histone modifications serve as an interface for environmental stimuli to exert control over the genome. These modifications have the capacity to activate or repress gene expression, thereby shaping the developmental outcomes of cells, circuits, and structures in the brain. Sex differences in methylation, methyl-binding proteins, and chromatin modifications indicate that epigenetic mechanism may be important for sexual differentiation of the brain. The data outlined in this review provide evidence that gonadal hormones in the neonatal brain influence epigenetic processes such as DNA methylation and histone acetylation, but also emphasize the primitive status of our current understanding of epigenetics and sexual differentiation and the brain.
Collapse
Affiliation(s)
- Bridget M Nugent
- Program in Neuroscience, University of Maryland, School of Medicine, Baltimore, MD, USA.
| | | |
Collapse
|
40
|
Analysis of estrogen receptor isoforms and variants in breast cancer cell lines. Exp Ther Med 2011; 2:537-544. [PMID: 22977537 DOI: 10.3892/etm.2011.226] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2010] [Accepted: 02/14/2011] [Indexed: 11/05/2022] Open
Abstract
In the present study, the expression of estrogen receptor (ER)α and ERβ isoforms in ER-positive (MCF7, T-47D and ZR-75-1) and ER-negative (MDA-MB-231, SK-BR-3, MDA-MB-453 and HCC1954) breast cancer cell lines was investigated. ERα mRNA was expressed in ER-positive and some ER-negative cell lines. ERα Δ3, Δ5 and Δ7 spliced variants were present in MCF7 and T-47D cells; ERα Δ5 and Δ7 spliced variants were detected in ZR-75-1 cells. MDA-MB-231 and HCC1954 cells expressed ERα Δ5 and Δ7 spliced variants. The ERβ1 variant was expressed in all of the cell lines and the ERβ2 variant in all of the ER-positive and some ER-negative cell lines (MDA-MB-231, MDA-MB-453 and SK-BR-3). MCF7, ZR-75-1, MDA-MB-453, HCC1954 and T-47D cells expressed ERβ5. All cell lines expressed an ERα 66-kDa protein band, and some expressed the truncated 42-kDa variant. ERβ1 was detected in all of the cell lines in addition to a 38-44 kDa variant. The results indicate that breast cancer cell lines widely used in research and reported as being ER-negative express ERα and/or ERβ mRNA and protein.
Collapse
|
41
|
Wilson ME, Westberry JM, Trout AL. Estrogen receptor-alpha gene expression in the cortex: sex differences during development and in adulthood. Horm Behav 2011; 59:353-7. [PMID: 20713055 PMCID: PMC3016448 DOI: 10.1016/j.yhbeh.2010.08.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Revised: 07/21/2010] [Accepted: 08/08/2010] [Indexed: 01/30/2023]
Abstract
17β-estradiol is a hormone with far-reaching organizational, activational and protective actions in both male and female brains. The organizational effects of early estrogen exposure are essential for long-lasting behavioral and cognitive functions. Estradiol mediates many of its effects through the intracellular receptors, estrogen receptor-alpha (ERα) and estrogen receptor-beta (ERβ). In the rodent cerebral cortex, estrogen receptor expression is high early in postnatal life and declines dramatically as the animal approaches puberty. This decline is accompanied by decreased expression of ERα mRNA. This change in expression is the same in both males and females in the developing isocortex and hippocampus. An understanding of the molecular mechanisms involved in the regulation of estrogen receptor alpha (ERα) gene expression is critical for understanding the developmental, as well as changes in postpubertal expression of the estrogen receptor. One mechanism of suppressing gene expression is by the epigenetic modification of the promoter regions by DNA methylation that results in gene silencing. The decrease in ERα mRNA expression during development is accompanied by an increase in promoter methylation. Another example of regulation of ERα gene expression in the adult cortex is the changes that occur following neuronal injury. Many animal studies have demonstrated that the endogenous estrogen, 17β-estradiol, is neuroprotective. Specifically, low levels of estradiol protect the cortex from neuronal death following middle cerebral artery occlusion (MCAO). In females, this protection is mediated through an ERα-dependent mechanism. ERα expression is rapidly increased following MCAO in females, but not in males. This increase is accompanied by a decrease in methylation of the promoter suggesting a return to the developmental program of gene expression within neurons. Taken together, during development and in adulthood, regulation of ERα gene expression in the cortex can occur by DNA methylation and in a sex-dependent fashion in the adult brain.
Collapse
Affiliation(s)
- Melinda E Wilson
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| | | | | |
Collapse
|
42
|
Heinzlmann A, Köves K, Kovács M, Csernus V. Sexual dimorphism in the effect of concomitant progesterone administration on changes caused by long-term estrogen treatment in pituitary hormone immunoreactivities of rats. Med Sci Monit 2011; 17:BR62-73. [PMID: 21358595 PMCID: PMC3524720 DOI: 10.12659/msm.881440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Accepted: 02/09/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Since in clinical practice long-term estrogen (E) treatment is frequently applied, our aim was to study the effect of concomitant progesterone (P) administration on changes caused by long-term estrogen treatment in the secretion of LH, FSH, PRL and GH. MATERIAL/METHODS Diethylstilbestrol (DES), P or both in silastic capsules were implanted under the skin of prepubertal Sprague-Dawley male and female rats. Animals survived for two or five months. We have also studied whether the changed hormone secretion caused by DES can return to normal level 1 or 2 months after removing DES capsule. RESULTS 1.) The males more rapidly responded than females with decreasing basal LH release upon treatments. The basal FSH release was decreased only in males. The effect of DES persisted in males; however, in females basal LH and FSH levels were upregulated after removal of DES capsule. 2.) The basal GH levels were low in each group. The body weight and length were depressed by DES in both genders and P little blunted this effect. The body weight and length in males remained low after removal of DES capsule, in females it was nearly similar to intact rats. 3.) There was no sexual dimorphism in the effect of steroids on PRL secretion. In both genders DES extremely enhanced the PRL levels, P prevented the effect of DES. PRL levels returned to intact value after removal of DES influence. 4.) Removal of DES capsule reversed the changes in the immunohistochemical appearance of hormone immunoreactivities. CONCLUSIONS There was sexual dimorphism in the change of basal gonadotropic hormone and GH secretion but not of PRL upon DES and DES+P treatments. P was basically protective and this role may be mediated by P receptors locally in the pituitary gland.
Collapse
Affiliation(s)
- Andrea Heinzlmann
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Katalin Köves
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | | | - Valér Csernus
- Department of Human Anatomy, Pécs University, Pécs, Hungary
| |
Collapse
|
43
|
Boulware MI, Kent BA, Frick KM. The impact of age-related ovarian hormone loss on cognitive and neural function. Curr Top Behav Neurosci 2011; 10:165-84. [PMID: 21533680 DOI: 10.1007/7854_2011_122] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
On average, women now live one-third of their lives after menopause. Because menopause has been associated with an elevated risk of dementia, an increasing body of research has studied the effects of reproductive senescence on cognitive function. Compelling evidence from humans, nonhuman primates, and rodents suggests that ovarian sex-steroid hormones can have rapid and profound effects on memory, attention, and executive function, and on regions of the brain that mediate these processes, such as the hippocampus and prefrontal cortex. This chapter will provide an overview of studies in humans, nonhuman primates, and rodents that examine the effects of ovarian hormone loss and hormone replacement on cognitive functions mediated by the hippocampus and prefrontal cortex. For humans and each animal model, we outline the effects of aging on reproductive function, describe how ovarian hormones (primarily estrogens) modulate hippocampal and prefrontal physiology, and discuss the effects of both reproductive aging and hormone treatment on cognitive function. Although this review will show that much has been learned about the effects of reproductive senescence on cognition, many critical questions remain for future investigation.
Collapse
Affiliation(s)
- Marissa I Boulware
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 E. Hartford Ave, Milwaukee, WI 53211, USA
| | | | | |
Collapse
|
44
|
Influence of different estrogens on neuroplasticity and cognition in the hippocampus. Biochim Biophys Acta Gen Subj 2010; 1800:1056-67. [DOI: 10.1016/j.bbagen.2010.01.006] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 01/13/2010] [Accepted: 01/16/2010] [Indexed: 11/18/2022]
|
45
|
Heinzlmann A, Köves K, Nagy GM. Effect of concomitant progesterone administration or the effect of removal of estrogen capsule on changes caused by long-term estrogen treatment in pituitary VIP immunoreactivities. Endocrine 2010; 37:396-402. [PMID: 20960159 DOI: 10.1007/s12020-010-9320-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
In the anterior pituitary besides the classical tropic hormones, peptides of a small molecular weight are also synthesized. One of them is the vasoactive intestinal polypeptide (VIP). VIP immunoreactivity is readily detected in human and monkey pituitaries; however, in the rat VIP immunoreactive cells were observed in about 50% of intact rats. In estrogen treated rats VIP immunoreactive cells were observed in the anterior pituitary of all animals. In this work, we have examined the effect of long-term sexual steroid treatments on the VIP immunoreactivity of the anterior pituitary using diethylstilbestrol (DES) or progesterone (P) filled capsules. The effectiveness of steroid treatments was tested by the measurement of plasma prolactin (PRL) level and by the appearance of prolactinoma. DES enhanced the plasma PRL level and 5 months later it induced prolactinomas, the concomitant P treatment prevented both the elevation of plasma PRL level and the formation of prolactinomas. These results indicated that there was enough steroid in the capsules. There was a positive correlation between the duration of DES influence and the number of VIP immunoreactive cells. Two months after the implantation of DES there was a considerable number of VIP cells in the anterior pituitary, and 5 months after implantation the number of VIP cells was greatly increased so as to form a VIP-oma. Concomitant implantation of P prevented the formation of VIP-oma. Two months after the implantation, the DES capsule was removed. Already 2 months after removal the number of VIP cells approximated to the control level. It has been concluded that P can prevent the undesired effect of DES not only on the PRL, but on the VIP immunoreactivity as well.
Collapse
Affiliation(s)
- Andrea Heinzlmann
- Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Tűzoltó u. 58, Budapest 1094, Hungary
| | | | | |
Collapse
|
46
|
Dadsetan M, Szatkowski JP, Shogren KL, Yaszemski MJ, Maran A. Hydrogel-mediated DNA delivery confers estrogenic response in nonresponsive osteoblast cells. J Biomed Mater Res A 2010; 91:1170-7. [PMID: 19148929 DOI: 10.1002/jbm.a.32291] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Oligo(polyethylene glycol) fumarate (OPF) hydrogel has been employed in musculoskeletal tissue engineering for photoencapsulation of chondrocytes and as a matrix for marrow stromal cells differentiation. In this study, we have studied the application of OPF hydrogel for coencapsulation of DNA and bone cells and examined whether coencapsulation can enhance gene transfer by maintaining the DNA within the cellular microenvironment. Our results showed that plasmid DNA encoding green fluorescence protein (GFP), coencapsulated with bone tumor cells, was capable of transfecting the cells, and the transfected tumor cells continuously expressed GFP protein over the time course of study (21 days). Furthermore, we have examined the coencapsulation of estrogen receptor (ER) encoding plasmid DNA and human fetal osteoblast cells (hFOB) that lack endogenous ER. Our results show that the transfected cells responded to estrogen as alkaline phosphatase (ALP), and estrogen response element (ERE)-directed luciferase enzyme activities increased with estrogen treatment. Taken together, these studies show that OPF hydrogel could be further explored for targeted gene delivery in bone and other tissues encapsulated within the hydrogels.
Collapse
Affiliation(s)
- M Dadsetan
- Department of Orthopedics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | |
Collapse
|
47
|
Ishii H, Kobayashi M, Sakuma Y. Alternative promoter usage and alternative splicing of the rat estrogen receptor alpha gene generate numerous mRNA variants with distinct 5'-ends. J Steroid Biochem Mol Biol 2010; 118:59-69. [PMID: 19833204 DOI: 10.1016/j.jsbmb.2009.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/26/2009] [Accepted: 10/04/2009] [Indexed: 11/29/2022]
Abstract
The 5'-untranslated region (UTR) of the estrogen receptor alpha (ERalpha) gene plays an important role in determining tissue-specific expression. To elucidate the regulatory mechanisms of rat ERalpha gene expression, the genomic organization must be investigated. We therefore analyzed the structure of the rat ERalpha mRNA 5'-UTR using rapid amplification of 5'-cDNA ends (5'-RACE) and RT-PCR. The analysis showed the presence of multiple variants containing unique 5'-UTRs. We mapped the cDNA sequences on the rat genome, and newly identified one leader exon (exon 0U) and ten untranslated internal exons (exons I1-10). Both splicing from four different leader exons (exons 0S, 0N, 0U, and 0/B) onto exon 1 and alternative splicing in combination with eleven internal exons (exons I1-10, and 0T) produce multiple transcripts. RT-PCR analysis revealed that each variant had preferred expression sites, suggesting that promoter usage and splicing are regulated in tissue-specific manners. Moreover, we determined a splicing event to yield Deltaexon 1 variants (0S-2-3-4-5-6-7-8), which are translated into rat 46 kDa ERalpha proteins. Our results indicate that the rat ERalpha gene is more complex than previously thought in terms of genomic organization and that both alternative promoter usage and alternative splicing contribute to the remarkable diversity of ERalpha mRNAs.
Collapse
Affiliation(s)
- Hirotaka Ishii
- Department of Physiology, Nippon Medical School, Tokyo, Japan.
| | | | | |
Collapse
|
48
|
Bopassa JC, Eghbali M, Toro L, Stefani E. A novel estrogen receptor GPER inhibits mitochondria permeability transition pore opening and protects the heart against ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2009; 298:H16-23. [PMID: 19880667 DOI: 10.1152/ajpheart.00588.2009] [Citation(s) in RCA: 143] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several studies have recently demonstrated that G protein-coupled receptor 30 (GPER) can directly bind to estrogen and mediate its action. We investigated the role and the mechanism of estrogen-induced cardioprotection after ischemia-reperfusion using a specific GPER agonist G1. Isolated hearts from male mice were perfused using Langendorff technique with oxygenated (95% O(2) and 5% CO(2)) Krebs Henseleit buffer (control), with G1 (1 microM), and G1 (1 microM) together with extracellular signal-regulated kinase (Erk) inhibitor PD-98059 (5 microM). After 20 min of perfusion, hearts were subjected to 20 min global normothermic (37 degrees C) ischemia followed by 40 min reperfusion. Cardiac function was measured, and myocardial necrosis was evaluated by triphenyltetrazolium chloride staining at the end of the reperfusion. Mitochondria were isolated after 10 min of reperfusion to assess the Ca(2+) load required to induce mitochondria permeability transition pore (mPTP) opening. G1-treated hearts developed better functional recovery with higher rate pressure product (RPP, 6140 +/- 264 vs. 2,640 +/- 334 beats mmHg(-1) min(-1), P < 0.05). The infarct size decreased significantly in G1-treated hearts (21 +/- 2 vs. 46 +/- 3%, P < 0.001), and the Ca(2+) load required to induce mPTP opening increased (2.4 +/- 0.06 vs. 1.6 +/- 0.11 microM/mg mitochondrial protein, P < 0.05) compared with the controls. The protective effect of G1 was abolished in the presence of PD-98059 [RPP: 4,120 +/- 46 beats mmHg(-1) min(-1), infarct size: 53 +/- 2%, and Ca(2+) retention capacity: 1.4 +/- 0.11 microM/mg mitochondrial protein (P < 0.05)]. These results suggest that GPER activation provides a cardioprotective effect after ischemia-reperfusion by inhibiting the mPTP opening, and this effect is mediated by the Erk pathway.
Collapse
Affiliation(s)
- Jean Chrisostome Bopassa
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095-7115, USA.
| | | | | | | |
Collapse
|
49
|
Boulware MI, Mermelstein PG. Membrane estrogen receptors activate metabotropic glutamate receptors to influence nervous system physiology. Steroids 2009; 74:608-13. [PMID: 19070630 PMCID: PMC2799184 DOI: 10.1016/j.steroids.2008.11.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2008] [Revised: 11/06/2008] [Accepted: 11/07/2008] [Indexed: 11/21/2022]
Abstract
Until recently, the idea that estradiol could affect cellular processes independent of nuclear estrogen receptors was often dismissed as artifact. This in spite of a large number of carefully controlled studies performed both within and outside the nervous system demonstrating estrogens regulate various intracellular signaling pathways by acting at the membrane surface of cells and/or at biological rates incompatible with the time course of genomic-initiated events. The concept that estradiol can act on surface membrane receptors to regulate nervous system function is now far less controversial. However, there is evidence that there may be multiple types of estrogen receptors on the membrane surface of cells. Determining the physiological relevance of each of these receptors is currently underway. Two important membrane estrogen receptors are in fact the classical estrogen receptor-alpha (ERalpha) and estrogen receptor-beta (ERbeta) proteins, which is somewhat surprising based upon their well-established role in nuclear gene transcription. This review will focus on the mechanism by which surface-localized ERalpha and ERbeta stimulate intracellular signaling events in cells of the nervous system through activation of metabotropic glutamate receptors (mGluRs). This mechanism of estrogen receptor function also requires caveolin proteins, which provide the subcellular compartmentalization of the particular signaling components required for appropriate cell stimulation. The review will conclude with several examples of physiological processes under the apparent regulation of ER/mGluR signaling.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Paul G. Mermelstein
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| |
Collapse
|
50
|
Barha CK, Lieblich SE, Galea LAM. Different forms of oestrogen rapidly upregulate cell proliferation in the dentate gyrus of adult female rats. J Neuroendocrinol 2009; 21:155-66. [PMID: 19076272 DOI: 10.1111/j.1365-2826.2008.01809.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oestrogens are known to exert significant structural and functional effects in the hippocampus of adult rodents. The dentate gyrus of the hippocampus retains the ability to produce neurones throughout adulthood and 17beta-oestradiol has been shown to influence hippocampal neurogenesis in adult female rats. The effects of other oestrogens, such as oestrone and 17alpha-oestradiol, on neurogenesis have not been investigated. The present study aimed to investigate the effects of 17beta-oestradiol, oestradiol benzoate, oestrone, and 17alpha-oestradiol on cell proliferation in ovariectomised adult female rats at two different time points. Young ovariectomised female rats were injected with one of the oestrogens at one of three doses. In Experiment 1, rats were exposed to the hormone for 4 h and, in Experiment 2, rats were exposed to the hormone for 30 min prior to 5-bromo-2-deoxyuridine injection to label proliferating cells and their progeny. We found that young ovariectomised females responded with increased cell proliferation to most oestrogens, except oestradiol benzoate, after 30 min of exposure. However, administration of oestrogens for a longer time interval was ineffective at increasing cell proliferation. After 30 min, 17beta-oestradiol and oestrone increased cell proliferation at low (0.3 microg) and high (10 microg) doses, whereas 17alpha-oestradiol increased cell proliferation at medium (1 microg) and high doses. The results of the present study indicate that different oestrogens rapidly increase cell proliferation in a dose-dependent manner, possibly through a nonclassical, nongenomic mechanism. Future experiments should focus on further elucidating the specific pathways utilised by each oestrogen. These results have important therapeutic implications because it may be possible to use 17alpha-oestradiol and lower doses of oestrogens in hormone replacement therapies.
Collapse
Affiliation(s)
- C K Barha
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | | | | |
Collapse
|