1
|
Camon C, Garratt M, Correa SM. Exploring the effects of estrogen deficiency and aging on organismal homeostasis during menopause. NATURE AGING 2024; 4:1731-1744. [PMID: 39672893 PMCID: PMC11785355 DOI: 10.1038/s43587-024-00767-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/28/2024] [Indexed: 12/15/2024]
Abstract
Sex hormone signaling declines during aging, from early midlife through menopause, as a consequence of reduced circulating estrogens and decreased receptiveness to these hormones in target tissues. Estrogens preserve energy homeostasis and promote metabolic health via coordinated and simultaneous effects throughout the brain and body. Age-associated loss of estrogen production during menopause has been implicated in a higher risk for metabolic diseases and increased mortality. However, it remains unclear whether age-associated changes in homeostasis are dependent on reduced estrogen signaling during menopause. Although menopausal hormone therapies containing estrogens can alleviate symptoms, concerns about the risks involved have contributed to a broad decline in the use of these approaches. Non-hormonal therapies have emerged that target tissues or pathways with varying levels of selectivity, reducing risk. We summarize here the broad effects of estrogen loss on homeostasis during menopause, current and emerging therapies and opportunities for understanding homeostatic disruptions associated with menopause.
Collapse
Affiliation(s)
- Celine Camon
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Garratt
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand.
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Wang R, Bai X, Yang H, Ma J, Yu S, Lu Z. Identification of a novel AIFM1 variant from a Chinese family with auditory neuropathy. Front Genet 2022; 13:1064823. [DOI: 10.3389/fgene.2022.1064823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Auditory neuropathy (AN) is a specific type of hearing loss characterized by impaired language comprehension. Apoptosis inducing factor mitochondrion associated 1 (AIFM1) is the most common gene associated with late-onset AN. In this study, we aimed to screen the pathogenic variant of AIFM1 in a Chinese family with AN and to explore the molecular mechanism underlying the function of such variant in the development of AN.Methods: One patient with AN and eight unaffected individuals from a Chinese family were enrolled in this study. A comprehensive clinical evaluation was performed on all participants. A targeted next-generation sequencing (NGS) analysis of a total of 406 known deafness genes was performed to screen the potential pathogenic variants in the proband. Sanger sequencing was used to confirm the variants identified in all participants. The pathogenicity of variant was predicted by bioinformatics analysis. Immunofluorescence and Western blot analyses were performed to evaluate the subcellular distribution and expression of the wild type (WT) and mutant AIFM1 proteins. Cell apoptosis was evaluated based on the TUNEL analyses.Results: Based on the clinical evaluations, the proband in this family was diagnosed with AN. The results of NGS and Sanger sequencing showed that a novel missense mutation of AIFM1, i.e., c.1367A > G (p. D456G), was identified in this family. Bioinformatics analysis indicated that this variant was pathogenic. Functional analysis showed that in comparison with the WT, the mutation c.1367A > G of AIFM1 showed no effect on its subcellular localization and the ability to induce apoptosis, but changed its protein expression level.Conclusion: A novel variant of AIFM1 was identified for the first time, which was probably the genetic cause of AN in a Chinese family with AN.
Collapse
|
3
|
Stincic TL, Kelly MJ. Estrogenic regulation of reproduction and energy homeostasis by a triumvirate of hypothalamic arcuate neurons. J Neuroendocrinol 2022; 34:e13145. [PMID: 35581942 DOI: 10.1111/jne.13145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17β-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
4
|
Estrogenic Action in Stress-Induced Neuroendocrine Regulation of Energy Homeostasis. Cells 2022; 11:cells11050879. [PMID: 35269500 PMCID: PMC8909319 DOI: 10.3390/cells11050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 01/27/2023] Open
Abstract
Estrogens are among important contributing factors to many sex differences in neuroendocrine regulation of energy homeostasis induced by stress. Research in this field is warranted since chronic stress-related psychiatric and metabolic disturbances continue to be top health concerns, and sex differences are witnessed in these aspects. For example, chronic stress disrupts energy homeostasis, leading to negative consequences in the regulation of emotion and metabolism. Females are known to be more vulnerable to the psychological consequences of stress, such as depression and anxiety, whereas males are more vulnerable to the metabolic consequences of stress. Sex differences that exist in the susceptibility to various stress-induced disorders have led researchers to hypothesize that gonadal hormones are regulatory factors that should be considered in stress studies. Further, estrogens are heavily recognized for their protective effects on metabolic dysregulation, such as anti-obesogenic and glucose-sensing effects. Perturbations to energy homeostasis using laboratory rodents, such as physiological stress or over-/under- feeding dietary regimen prevalent in today’s society, offer hints to the underlying mechanisms of estrogenic actions. Metabolic effects of estrogens primarily work through estrogen receptor α (ERα), which is differentially expressed between the sexes in hypothalamic nuclei regulating energy metabolism and in extrahypothalamic limbic regions that are not typically associated with energy homeostasis. In this review, we discuss estrogenic actions implicated in stress-induced sex-distinct metabolic disorders.
Collapse
|
5
|
Mohr MA, Wong AM, Sukumar G, Dalgard CL, Hong W, Wu TJ, Wu YE, Micevych PE. RNA-sequencing of AVPV and ARH reveals vastly different temporal and transcriptomic responses to estradiol in the female rat hypothalamus. PLoS One 2021; 16:e0256148. [PMID: 34407144 PMCID: PMC8372949 DOI: 10.1371/journal.pone.0256148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/30/2021] [Indexed: 11/18/2022] Open
Abstract
In females, estrogens have two main modes of action relating to gonadotropin secretion: positive feedback and negative feedback. Estrogen positive and negative feedback are controlled by different regions of the hypothalamus: the preoptic area/anterior portion (mainly the anteroventral periventricular nucleus, AVPV) of the hypothalamus is associated with estrogen positive feedback while the mediobasal hypothalamus (mainly the arcuate nucleus of the hypothalamus, ARH), is associated with estrogen negative feedback. In this study, we examined the temporal pattern of gene transcription in these two regions following estrogen treatment. Adult, ovariectomized, Long Evans rats received doses of estradiol benzoate (EB) or oil every 4 days for 3 cycles. On the last EB priming cycle, hypothalamic tissues were dissected into the AVPV+ and ARH+ at 0 hrs (baseline/oil control), 6 hrs, or 24 hrs after EB treatment. RNA was extracted and sequenced using bulk RNA sequencing. Differential gene analysis, gene ontology, and weighted correlation network analysis (WGCNA) was performed. Overall, we found that the AVPV+ and ARH+ respond differently to estradiol stimulation. In both regions, estradiol treatment resulted in more gene up-regulation than down-regulation. S100g was very strongly up-regulated by estradiol in both regions at 6 and 24 hrs after EB treatment. In the AVPV+ the highest number of differentially expressed genes occurred 24 hrs after EB. In the ARH+, the highest number of genes differentially expressed by EB occurred between 6 and 24 hrs after EB, while in the AVPV+, the fewest genes changed their expression between these time points, demonstrating a temporal difference in the way that EB regulates transcription these two areas. Several genes strongly implicated in gonadotropin release were differentially affected by estradiol including Esr1, encoding estrogen receptor-α and Kiss1, encoding kisspeptin. As an internal validation, Kiss1 was up-regulated in the AVPV+ and down-regulated in the ARH+. Gene network analysis revealed the vastly different clustering of genes modulated by estradiol in the AVPV+ compared with the ARH+. These results indicate that gene expression in these two hypothalamic regions have specific responses to estradiol in timing and direction.
Collapse
Affiliation(s)
- Margaret A. Mohr
- Dept of Neurobiology, and the Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA DGSOM, Los Angeles, California, United States of America
| | - Angela M. Wong
- Dept of Neurobiology, and the Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA DGSOM, Los Angeles, California, United States of America
| | - Gauthaman Sukumar
- Dept of Gynecological Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Clifton L. Dalgard
- Dept of Gynecological Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Weizhe Hong
- Dept of Neurobiology, and the Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA DGSOM, Los Angeles, California, United States of America
| | - T. John Wu
- Dept of Gynecological Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Ye Emily Wu
- Dept of Neurobiology, and the Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA DGSOM, Los Angeles, California, United States of America
| | - Paul E. Micevych
- Dept of Neurobiology, and the Laboratory of Neuroendocrinology of the Brain Research Institute, UCLA DGSOM, Los Angeles, California, United States of America
| |
Collapse
|
6
|
Stincic TL, Rønnekleiv OK, Kelly MJ. Membrane and nuclear initiated estrogenic regulation of homeostasis. Steroids 2021; 168:108428. [PMID: 31229508 PMCID: PMC6923613 DOI: 10.1016/j.steroids.2019.108428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/08/2019] [Accepted: 06/18/2019] [Indexed: 11/23/2022]
Abstract
Reproduction and energy balance are inextricably linked in order to optimize the evolutionary fitness of an organism. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy and produce unhealthy or obesity-prone offspring. The quintessential function of the hypothalamus is to act as a bridge between the endocrine and nervous systems, coordinating fertility and autonomic functions. Across the female reproductive cycle various motivations wax and wane, following levels of ovarian hormones. Estrogens, more specifically 17β-estradiol (E2), coordinate a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool of cells, this triumvirate is composed of the kisspeptin (Kiss1ARH), proopiomelanocortin (POMC), and neuropeptide Y/agouti-related peptide (AgRP) neurons. Although the excitability of these neuronal subpopulations is subject to genomic and rapid estrogenic regulation, kisspeptin neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we will review the recent findings on the synaptic interactions between Kiss1, AgRP and POMC neurons and how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, United States
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, United States; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR 97006, United States.
| |
Collapse
|
7
|
Assessing the Anti-inflammatory Mechanism of Reduning Injection by Network Pharmacology. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6134098. [PMID: 33381562 PMCID: PMC7758122 DOI: 10.1155/2020/6134098] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/30/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022]
Abstract
Reduning Injection (RDNI) is a traditional Chinese medicine formula indicated for the treatment of inflammatory diseases. However, the molecular mechanism of RDNI is unclear. The information of RDNI ingredients was collected from previous studies. Targets of them were obtained by data mining and molecular docking. The information of targets and related pathways was collected in UniProt and KEGG. Networks were constructed and analyzed by Cytoscape to identify key compounds, targets, and pathways. Data mining and molecular docking identified 11 compounds, 84 targets, and 201 pathways that are related to the anti-inflammatory activity of RDNI. Network analysis identified two key compounds (caffeic acid and ferulic acid), five key targets (Bcl-2, eNOS, PTGS2, PPARA, and MMPs), and four key pathways (estrogen signaling pathway, PI3K-AKT signaling pathway, cGMP-PKG signaling pathway, and calcium signaling pathway) which would play critical roles in the treatment of inflammatory diseases by RDNI. The cross-talks among pathways provided a deeper understanding of anti-inflammatory effect of RDNI. RDNI is capable of regulating multiple biological processes and treating inflammation at a systems level. Network pharmacology is a practical approach to explore the therapeutic mechanism of TCM for complex disease.
Collapse
|
8
|
Vail GM, Roepke TA. Organophosphate Flame Retardants Excite Arcuate Melanocortin Circuitry and Increase Neuronal Sensitivity to Ghrelin in Adult Mice. Endocrinology 2020; 161:5910086. [PMID: 32961558 PMCID: PMC7575050 DOI: 10.1210/endocr/bqaa168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/17/2020] [Indexed: 12/28/2022]
Abstract
Organophosphate flame retardants (OPFRs) are a class of chemicals that have become near ubiquitous in the modern environment. While OPFRs provide valuable protection against flammability of household items, they are increasingly implicated as an endocrine disrupting chemical (EDC). We previously reported that exposure to a mixture of OPFRs causes sex-dependent disruptions of energy homeostasis through alterations in ingestive behavior and activity in adult mice. Because feeding behavior and energy expenditure are largely coordinated by the hypothalamus, we hypothesized that OPFR disruption of energy homeostasis may occur through EDC action on melanocortin circuitry within the arcuate nucleus. To this end, we exposed male and female transgenic mice expressing green fluorescent protein in either neuropeptide Y (NPY) or proopiomelanocortin (POMC) neurons to a common mixture of OPFRs (triphenyl phosphate, tricresyl phosphate, and tris(1,3-dichloro-2-propyl)phosphate; each 1 mg/kg bodyweight/day) for 4 weeks. We then electrophysiologically examined neuronal properties using whole-cell patch clamp technique. OPFR exposure depolarized the resting membrane of NPY neurons and dampened a hyperpolarizing K+ current known as the M-current within the same neurons from female mice. These neurons were further demonstrated to have increased sensitivity to ghrelin excitation, which more potently reduced the M-current in OPFR-exposed females. POMC neurons from female mice exhibited elevated baseline excitability and are indicated in receiving greater excitatory synaptic input when exposed to OPFRs. Together, these data support a sex-selective effect of OPFRs to increase neuronal output from the melanocortin circuitry governing feeding behavior and energy expenditure, and give reason for further examination of OPFR impact on human health.
Collapse
Affiliation(s)
- Gwyndolin M Vail
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Troy A Roepke
- Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Department of Animal Sciences, School of Environmental & Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
- Environmental and Occupational Health Science Institute, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Rutgers Center for Lipid Research, Center for Nutrition, Microbiome, and Health, and New Jersey Institute of Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
9
|
The loss of ERE-dependent ERα signaling potentiates the effects of maternal high-fat diet on energy homeostasis in female offspring fed an obesogenic diet. J Dev Orig Health Dis 2019; 11:285-296. [PMID: 31543088 DOI: 10.1017/s2040174419000515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal high-fat diet (HFD) alters hypothalamic programming and disrupts offspring energy homeostasis in rodents. We previously reported that the loss of ERα signaling partially blocks the effects of maternal HFD in female offspring fed a standard chow diet. In a companion study, we determined if the effects of maternal HFD were magnified by an adult obesogenic diet in our transgenic mouse models. Heterozygous ERα knockout (wild-type (WT)/KO) dams were fed a control breeder chow diet (25% fat) or a semipurified HFD (45% fat) 4 weeks prior to mating with heterozygous males (WT/KO or WT/ knockin) to produce WT, ERα KO, or ERα knockin/knockout (KIKO) (no estrogen response element (ERE) binding) female offspring, which were fed HFD for 20 weeks. Maternal HFD potentiated the effects of adult HFD on KIKO and KO body weight due to increased adiposity and decreased activity. Maternal HFD also produced KIKO females that exhibit KO-like insulin intolerance and impaired glucose homeostasis. Maternal HFD increased plasma interleukin 6 and monocyte chemoattractant protein 1 levels and G6pc and Pepck liver expression only in WT mice. Insulin and tumor necrosis factor α levels were higher in KO offspring from HFD-fed dams. Arcuate and liver expression of Esr1 was altered in KIKO and WT, respectively. These data suggest that loss of ERE-dependent ERα signaling, and not total ERα signaling, sensitizes females to the deleterious influence of maternal HFD on offspring energy and glucose potentially through the control of peripheral inflammation and hypothalamic and liver gene expression. Future studies will interrogate the tissue-specific mechanisms of maternal HFD programming through ERα signaling.
Collapse
|
10
|
Qiu J, Bosch MA, Zhang C, Rønnekleiv OK, Kelly MJ. Estradiol Protects Neuropeptide Y/Agouti-Related Peptide Neurons against Insulin Resistance in Females. Neuroendocrinology 2019; 110:105-118. [PMID: 31212279 PMCID: PMC6920578 DOI: 10.1159/000501560] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/17/2019] [Indexed: 12/18/2022]
Abstract
When it comes to obesity, men exhibit a higher incidence of metabolic syndrome than women in early adult life, but this sex advantage wanes in postmenopausal women. A key diagnostic of the metabolic syndrome is insulin resistance in both peripheral tissues and brain, especially in the hypothalamus. Since the anorexigenic hormone 17β-estradiol (E2) regulates food intake in part by inhibiting the excitability of the hypothalamic neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons, we hypothesized that E2 would protect against insulin resistance in NPY/AgRP neurons with diet-induced obesity (DIO). Therefore, we did whole-cell recordings and single cell quantitative polymerase chain reaction in arcuate NPYGFP neurons from both female and male mice to test the efficacy of insulin with DIO. The resting membrane potential and input resistance of NPY/AgRP neurons were significantly increased in DIO versus control-diet fed males. Most notably, the efficacy of insulin to activate KATP channels in NPY/AgRP neurons was significantly attenuated, although the KATP channel opener diazoxide was fully effective in NPY/AgRP neurons from DIO males, indicating that the KATP channels were expressed and functional. In contrast, insulin was fully efficacious to activate KATP channels in DIO females, and the response was reversed by the KATP channel blocker tolbutamide. However, the ability of insulin to activate KATP channels was abrogated with ovariectomy but fully restored with E2 replacement. Insulin resistance in obese males was likely mediated by an increase in suppressor of cytokine signaling-3 (SOCS-3), protein tyrosine phosphatase B (PTP1B) and T-cell protein tyrosine phosphatase (TCPTP) activity, since the expression of all 3 mRNAs were upregulated in the obese males but not in females. As proof of principle, pre-incubation of hypothalamic slices from DIO males with the PTP1B/TCPTP inhibitor CX08005 completely rescued the effects of insulin. Therefore, E2 protects NPY/AgRP neurons in females against insulin resistance through, at least in part, attenuating phosphatase activity. The neuroprotective effects of E2 may explain sex differences in the expression of metabolic syndrome that disappears with the loss of E2 in aging.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Chunguang Zhang
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
- Division of Neuroscience, National Primate Research Center,
Oregon Health & Science University, Beaverton, Oregon, USA
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health
& Science University, Portland, Oregon, USA
- Division of Neuroscience, National Primate Research Center,
Oregon Health & Science University, Beaverton, Oregon, USA
| |
Collapse
|
11
|
Tran TKA, Yu RMK, Islam R, Nguyen THT, Bui TLH, Kong RYC, O'Connor WA, Leusch FDL, Andrew-Priestley M, MacFarlane GR. The utility of vitellogenin as a biomarker of estrogenic endocrine disrupting chemicals in molluscs. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 248:1067-1078. [PMID: 31091639 DOI: 10.1016/j.envpol.2019.02.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/31/2019] [Accepted: 02/18/2019] [Indexed: 06/09/2023]
Abstract
Estrogenic endocrine disrupting chemicals (EDCs) are natural hormones, synthetic compounds or industrial chemicals that mimic estrogens due to their structural similarity with estrogen's functional moieties. They typically enter aquatic environments through wastewater treatment plant effluents or runoff from intensive livestock operations. Globally, most natural and synthetic estrogens in receiving aquatic environments are in the low ng/L range, while industrial chemicals (such as bisphenol A, nonylphenol and octylphenol) are present in the μg to low mg/L range. These environmental concentrations often exceed laboratory-based predicted no effect concentrations (PNECs) and have been evidenced to cause negative reproductive impacts on resident aquatic biota. In vertebrates, such as fish, a well-established indicator of estrogen-mediated endocrine disruption is overexpression of the egg yolk protein precursor vitellogenin (Vtg) in males. Although the vertebrate Vtg has high sensitivity and specificity to estrogens, and the molecular basis of its estrogen inducibility has been well studied, there is growing ethical concern over the use of vertebrate animals for contaminant monitoring. The potential utility of the invertebrate Vtg as a biomonitor for environmental estrogens has therefore gained increasing attention. Here we review evidence providing support that the molluscan Vtg holds promise as an invertebrate biomarker for exposure to estrogens. Unlike vertebrates, estrogen signalling in invertebrates remains largely unclarified and the classical genomic pathway only partially explains estrogen-mediated activation of Vtg. In light of this, in the latter part of this review, we summarise recent progress towards understanding the molecular mechanisms underlying the activation of the molluscan Vtg gene by estrogens and present a hypothetical model of the interplay between genomic and non-genomic pathways in the transcriptional regulation of the gene.
Collapse
Affiliation(s)
- Thi Kim Anh Tran
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Institute for Agriculture and Resources, Vinh University, Viet Nam
| | - Richard Man Kit Yu
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Rafiquel Islam
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Department of Applied Chemistry and Chemical Engineering, Islamic University, Kushtia, 7003, Bangladesh
| | - Thi Hong Tham Nguyen
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Institute for Agriculture and Resources, Vinh University, Viet Nam
| | - Thi Lien Ha Bui
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia; Division of Experimental Biology, Research Institute for Aquaculture No 2, Viet Nam
| | - Richard Yuen Chong Kong
- Department of Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong Special Administrative Region, China
| | - Wayne A O'Connor
- New South Wales Department of Primary Industries, Port Stephens Fisheries Institute, Taylors Beach, NSW, 2316, Australia
| | - Frederic D L Leusch
- Australian Rivers Institute, Griffith School of Environment and Science, Griffith University, QLD, 4111, Australia
| | | | - Geoff R MacFarlane
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW, 2308, Australia.
| |
Collapse
|
12
|
Vail G, Roepke TA. Membrane-initiated estrogen signaling via Gq-coupled GPCR in the central nervous system. Steroids 2019; 142:77-83. [PMID: 29378226 PMCID: PMC6064680 DOI: 10.1016/j.steroids.2018.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 12/08/2017] [Accepted: 01/19/2018] [Indexed: 01/21/2023]
Abstract
The last few decades have revealed increasing complexity and depth to our knowledge of receptor-mediated estrogen signaling. Nuclear estrogen receptors (ERs) ERα and ERβ remain the fundamental dogma, but recent research targeting membrane-bound ERs urges for a more expanded view on ER signaling. ERα and ERβ are also involved in membrane-delineated signaling alongside membrane-specific G protein-coupled estrogen receptor 1 (GPER1), ER-X, and the Gq-coupled membrane ER (Gq-mER). Membrane ERs are responsible for eliciting rapid responses to estrogen signaling, and their importance has been increasingly indicated in central nervous system (CNS) regulation of such functions as reproduction, energy homeostasis, and stress. While the Gq-mER signaling pathway is well characterized, the receptor structure and gene remains uncharacterized, although it is not similar to the nuclear ERα/β. This review will describe the current knowledge of this putative membrane ER and its selective ligand, STX, from its initial characterization in hypothalamic melanocortin circuitry to recent research exploring its role in the CNS outside of the hypothalamus.
Collapse
Affiliation(s)
- Gwyndolin Vail
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States; Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States; Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States.
| |
Collapse
|
13
|
Estradiol Drives the Anorexigenic Activity of Proopiomelanocortin Neurons in Female Mice. eNeuro 2018; 5:eN-NWR-0103-18. [PMID: 30310864 PMCID: PMC6179576 DOI: 10.1523/eneuro.0103-18.2018] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/11/2018] [Accepted: 07/02/2018] [Indexed: 12/14/2022] Open
Abstract
Energy balance is regulated by anorexigenic proopiomelanocortin (POMC) and orexigenic neuropeptide Y/agouti-related peptide (NPY/AgRP) neurons of the hypothalamic arcuate nucleus. POMC neurons make extensive projections and are thought to release both amino acid and peptide neurotransmitters. However, whether they communicate directly with NPY/AgRP neurons is debated. Initially, using single-cell RT-PCR, we determined that mouse POMCeGFP neurons express Slc17a6 (Vglut2) and Slc18a2 (Vmat2), but not Slc31a1 (Vgat) mRNA, suggesting glutamate and non-canonical GABA release. Quantitative (q)RT-PCR of POMCeGFP cells revealed that Vglut2 and Vmat2 expression was significantly increased in E2- versus oil-treated, ovariectomized (OVX) female mice. Since 17β-estradiol (E2) is anorexigenic, we hypothesized that an underlying mechanism is enhancement of POMC signaling. Therefore, we optogenetically stimulated POMC neurons in hypothalamic slices to examine evoked release of neurotransmitters onto NPY/AgRP neurons. Using brief light pulses, we primarily observed glutamatergic currents and, based on the paired pulse ratio (PPR), determined that release probability was higher in E2- versus oil-treated, OVX female, congruent with increased Vlgut2 expression. Moreover, bath perfusion of the Gq-coupled membrane estrogen receptor (ER) agonist STX recapitulated the effects of E2 treatment. In addition, high-frequency (20 Hz) stimulation generated a slow outward current that reversed near Ek+ and was antagonized by naloxone, indicative of β-endorphin release. Furthermore, individual NPY/AgRP neurons were found to express Oprm1, the transcript for μ-opioid receptor, and DAMGO, a selective agonist, elicited an outward current. Therefore, POMC excitability and neurotransmission are enhanced by E2, which would facilitate decreased food consumption through marked inhibition of NPY/AgRP neurons.
Collapse
|
14
|
Song MY, Li CY, Liu XF, Xiao JY, Zhao H. Effect of 17β-oestradiol on T-type calcium channels in the lateral habenula. J Neuroendocrinol 2018; 30:e12629. [PMID: 29917292 DOI: 10.1111/jne.12629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Revised: 06/10/2018] [Accepted: 06/16/2018] [Indexed: 11/27/2022]
Abstract
T-type calcium channels (T-channels) are critical for regulating neuronal excitability. Oestrogen alters neuronal excitability by modulating the expression of T-channels. The lateral habenula (LHb), as a link between the limbic system and midbrain structures, expresses T-channels and ERs. However, little is known about the role of oestrogen with respect to modulating T-channels in the LHb. In the present study, we investigated the distribution of T-channels in 3 LHb subregions (rostral, middle and caudal) in normal female rats. Next, we analysed the influence of 17β-oestradiol (E2 ) on T-channels in the LHb in ovariectomised (OVX) rats (oil and E2 groups) using whole-cell patch clamp recording and a real-time polymerase chain reaction (PCR). In normal rats, the results obtained showed that the peak of T-type calcium current (IT ) was -474.61 ± 48.33 pA and IT density was -29.11 ± 1.93 pA/pF. The IT peak and IT density on LHb neurones gradually decreased across the rostrocaudal axis. The neuronal firing pattern varied depending on the location: burst firing was dominant (53.85%) in the rostral LHb, whereas tonic firing was dominant (79.31%) in the caudal LHb. In OVX rats, real-time PCR analysis revealed that E2 treatment decreased Cav3.3 mRNA expression in the caudal LHb. Patch clamp recording showed that E2 treatment decreased the peak IT and also reduced the low-threshold spikes (LTS) number, amplitude and width of LTS in the caudal LHb. Taken together, the results obtained in the present study suggest that E2 may inhibit T-channel activity by selectively down-regulating Cav3.3 calcium channel in the caudal LHb, leading to reduced the possibility of burst firing.
Collapse
Affiliation(s)
- Mei Ying Song
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chun Ying Li
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiao Feng Liu
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jin Yu Xiao
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Hua Zhao
- Neuroscience Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Physiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
15
|
Krolick KN, Zhu Q, Shi H. Effects of Estrogens on Central Nervous System Neurotransmission: Implications for Sex Differences in Mental Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:105-171. [PMID: 30470289 PMCID: PMC6737530 DOI: 10.1016/bs.pmbts.2018.07.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nearly one of every five US individuals aged 12 years old or older lives with certain types of mental disorders. Men are more likely to use various types of substances, while women tend to be more susceptible to mood disorders, addiction, and eating disorders, all of which are risks associated with suicidal attempts. Fundamental sex differences exist in multiple aspects of the functions and activities of neurotransmitter-mediated neural circuits in the central nervous system (CNS). Dysregulation of these neural circuits leads to various types of mental disorders. The potential mechanisms of sex differences in the CNS neural circuitry regulating mood, reward, and motivation are only beginning to be understood, although they have been largely attributed to the effects of sex hormones on CNS neurotransmission pathways. Understanding this topic is important for developing prevention and treatment of mental disorders that should be tailored differently for men and women. Studies using animal models have provided important insights into pathogenesis, mechanisms, and new therapeutic approaches of human diseases, but some concerns remain to be addressed. The purpose of this chapter is to integrate human and animal studies involving the effects of the sex hormones, estrogens, on CNS neurotransmission, reward processing, and associated mental disorders. We provide an overview of existing evidence for the physiological, behavioral, cellular, and molecular actions of estrogens in the context of controlling neurotransmission in the CNS circuits regulating mood, reward, and motivation and discuss related pathology that leads to mental disorders.
Collapse
Affiliation(s)
- Kristen N Krolick
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Qi Zhu
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States
| | - Haifei Shi
- Center for Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH, United States; Cellular, Molecular and Structural Biology, Miami University, Oxford, OH, United States.
| |
Collapse
|
16
|
Stincic TL, Rønnekleiv OK, Kelly MJ. Diverse actions of estradiol on anorexigenic and orexigenic hypothalamic arcuate neurons. Horm Behav 2018; 104:146-155. [PMID: 29626486 PMCID: PMC6196116 DOI: 10.1016/j.yhbeh.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/13/2022]
Abstract
Contribution to Special Issue on Fast effects of steroids. There is now compelling evidence for membrane-associated estrogen receptors in hypothalamic neurons that are critical for the hypothalamic control of homeostatic functions. It has been known for some time that estradiol (E2) can rapidly alter hypothalamic neuronal activity within seconds, indicating that some cellular effects can occur via membrane initiated events. However, our understanding of how E2 signals via membrane-associated receptors and how these signals impact physiological functions is only just emerging. Thus, E2 can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell excitability and even gene transcription in hypothalamic neurons. One population of hypothalamic neurons, the anorexigenic proopiomelanocortin (POMC) neurons, has long been considered to be a target of E2's actions based on gene (Pomc) expression studies. However, we now know that E2 can rapidly alter POMC neuronal activity within seconds and activate several intracellular signaling cascades that ultimately affect gene expression, actions which are critical for maintaining sensitivity to insulin in metabolically stressed states. E2 also affects the orexigenic Neuropeptide Y/Agouti-related Peptide (NPY/AgRP) neurons in similarly rapid but antagonistic manner. Therefore, this review will summarize our current state of knowledge of how E2 signals via rapid membrane-initiated and intracellular signaling cascades in POMC and NPY/AgRP neurons to regulate energy homeostasis.
Collapse
Affiliation(s)
- Todd L Stincic
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Oline K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Martin J Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
17
|
Gourdy P, Guillaume M, Fontaine C, Adlanmerini M, Montagner A, Laurell H, Lenfant F, Arnal JF. Estrogen receptor subcellular localization and cardiometabolism. Mol Metab 2018; 15:56-69. [PMID: 29807870 PMCID: PMC6066739 DOI: 10.1016/j.molmet.2018.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND In addition to their crucial role in reproduction, estrogens are key regulators of energy and glucose homeostasis and they also exert several cardiovascular protective effects. These beneficial actions are mainly mediated by estrogen receptor alpha (ERα), which is widely expressed in metabolic and vascular tissues. As a member of the nuclear receptor superfamily, ERα was primarily considered as a transcription factor that controls gene expression through the activation of its two activation functions (ERαAF-1 and ERαAF-2). However, besides these nuclear actions, a pool of ERα is localized in the vicinity of the plasma membrane, where it mediates rapid signaling effects called membrane-initiated steroid signals (MISS) that have been well described in vitro, especially in endothelial cells. SCOPE OF THE REVIEW This review aims to summarize our current knowledge of the mechanisms of nuclear vs membrane ERα activation that contribute to the cardiometabolic protection conferred by estrogens. Indeed, new transgenic mouse models (affecting either DNA binding, activation functions or membrane localization), together with the use of novel pharmacological tools that electively activate membrane ERα effects recently allowed to begin to unravel the different modes of ERα signaling in vivo. CONCLUSION Altogether, available data demonstrate the prominent role of ERα nuclear effects, and, more specifically, of ERαAF-2, in the preventive effects of estrogens against obesity, diabetes, and atheroma. However, membrane ERα signaling selectively mediates some of the estrogen endothelial/vascular effects (NO release, reendothelialization) and could also contribute to the regulation of energy balance, insulin sensitivity, and glucose metabolism. Such a dissection of ERα biological functions related to its subcellular localization will help to understand the mechanism of action of "old" ER modulators and to design new ones with an optimized benefit/risk profile.
Collapse
Affiliation(s)
- Pierre Gourdy
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service de Diabétologie, Maladies Métaboliques et Nutrition, CHU de Toulouse, Toulouse, France.
| | - Maeva Guillaume
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France; Service d'Hépatologie et Gastro-Entérologie, CHU de Toulouse, Toulouse, France
| | - Coralie Fontaine
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Marine Adlanmerini
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Alexandra Montagner
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Françoise Lenfant
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| | - Jean-François Arnal
- Institut des Maladies Métaboliques et Cardiovasculaires, UMR 1048/I2MC, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Toulouse, Toulouse, France
| |
Collapse
|
18
|
Qiu J, Bosch MA, Meza C, Navarro UV, Nestor CC, Wagner EJ, Rønnekleiv OK, Kelly MJ. Estradiol Protects Proopiomelanocortin Neurons Against Insulin Resistance. Endocrinology 2018; 159:647-664. [PMID: 29165691 PMCID: PMC5774249 DOI: 10.1210/en.2017-00793] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/01/2017] [Indexed: 12/24/2022]
Abstract
Insulin resistance is at the core of the metabolic syndrome, and men exhibit a higher incidence of metabolic syndrome than women in early adult life, but this sex advantage diminishes sharply when women reach the postmenopausal state. Because 17β-estradiol (E2) augments the excitability of the anorexigenic proopiomelanocortin (POMC) neurons, we investigated the neuroprotective effects of E2 against insulin resistance in POMC neurons from diet-induced obese (DIO) female and male mice. The efficacy of insulin to activate canonical transient receptor potential 5 (TRPC5) channels and depolarize POMC neurons was significantly reduced in DIO male mice but not in DIO female mice. However, the insulin response in POMC neurons was abrogated in ovariectomized DIO females but restored with E2 replacement. E2 increased T-type calcium channel Cav3.1 messenger RNA (mRNA) expression and whole-cell currents but downregulated stromal-interaction molecule 1 mRNA, which rendered POMC neurons more excitable and responsive to insulin-mediated TRPC5 channel activation. Moreover, E2 prevented the increase in suppressor of cytokine signaling-3 mRNA expression with DIO as seen in DIO males. As proof of principle, insulin [intracerebroventricular injection into the third ventricle (ICV)] decreased food intake and increased metabolism in female but not male guinea pigs fed a high-fat diet. The uncoupling of the insulin receptor from its downstream effector system was corroborated by the reduced expression of phosphorylated protein kinase B in the arcuate nucleus of male but not female guinea pigs following insulin. Therefore, E2 protects female POMC neurons from insulin resistance by enhancing POMC neuronal excitability and the coupling of insulin receptor to TRPC5 channel activation.
Collapse
Affiliation(s)
- Jian Qiu
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Cecilia Meza
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California 91766
| | - Uyen-Vy Navarro
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Casey C Nestor
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
| | - Edward J. Wagner
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific Western University of Health Sciences, Pomona, California 91766
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon 97006
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon 97239
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon 97006
| |
Collapse
|
19
|
Wehling M. Rapid actions of aldosterone revisited: Receptors in the limelight. J Steroid Biochem Mol Biol 2018; 176:94-98. [PMID: 28126566 DOI: 10.1016/j.jsbmb.2017.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 01/05/2017] [Accepted: 01/22/2017] [Indexed: 12/27/2022]
Abstract
Steroid hormones like aldosterone have been conclusively shown to elicit both late genomic and rapid, nongenomically initiated responses. Aldosterone was among the first for which rapid, clinically relevant effects were even shown in humans. Yet, after over 30 years of research, the nature of receptors involved in rapid actions of aldosterone is still unclear. Such effects may be assigned to the classical, intracellular steroid receptors, in this case mineralocorticoid receptors (MR, class IIa action Mannheim classification). They typically disappear in knockout models and are blocked by MR-antagonists such as spironolactone, as shown for several cellular and physiological, e.g. renal or cardiovascular effects. In contrast, there is also consistent evidence suggesting type IIb effects involving structurally different receptors ("membrane receptors") being insensitive to classic antagonists and persistent in knockout models; IIb effects have lately even been confirmed by atomic force detection of surface receptors which bind aldosterone but not spironolactone. Type IIa and b may coexist in the same cell with IIa often augmenting early IIb effects. So far cloning of IIb receptors was unsuccessful; therefore results on G-protein coupled estrogen receptor 1 (GPER1) being potentially involved in rapid aldosterone action raised considerable interest. Surprisingly, GPER1 does not bind aldosterone. Though under these circumstances GPER1 should not yet be considered as IIb-receptor, it might be an intermediary signaling enhancer of mineralocorticoid action as shown for epithelial growth factor receptors reconciling those results. We still seem to be left without IIb-receptors whose identification would however be highly desirable and essential for clinical translation.
Collapse
Affiliation(s)
- Martin Wehling
- University of Heidelberg, Clinical Pharmacology Mannheim, Theodor-Kutzer-Ufer 1-3, D-68167, Mannheim, Germany.
| |
Collapse
|
20
|
Roepke TA, Yasrebi A, Villalobos A, Krumm EA, Yang JA, Mamounis KJ. Loss of ERα partially reverses the effects of maternal high-fat diet on energy homeostasis in female mice. Sci Rep 2017; 7:6381. [PMID: 28743985 PMCID: PMC5526977 DOI: 10.1038/s41598-017-06560-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 06/13/2017] [Indexed: 11/18/2022] Open
Abstract
Maternal high-fat diet (HFD) alters hypothalamic developmental programming and disrupts offspring energy homeostasis in rodents. 17β-estradiol (E2) also influences hypothalamic programming through estrogen receptor (ER) α. Therefore, we hypothesized that females lacking ERα would be more susceptible to maternal HFD. To address this question, heterozygous ERα knockout (WT/KO) dams were fed a control breeder chow diet (25% fat) or a semi-purified HFD (45% fat) 4 weeks prior to mating with WT/KO males or heterozygous males with an ERα DNA-binding domain mutation knocked in (WT/KI) to produce WT, ERα KO, or ERα KIKO females lacking ERE-dependent ERα signaling. Maternal HFD increased body weight in WT and KIKO, in part, due to increased adiposity and daytime carbohydrate utilization in WT and KIKO, while increasing nighttime fat utilization in KO. Maternal HFD also increased plasma leptin, IL-6, and MCP-1 in WT and increased arcuate expression of Kiss1 and Esr1 (ERα) and liver expression of G6pc and Pepck in WT and KIKO. Contrary to our hypothesis, these data suggest that loss of ERα signaling blocks the influence of maternal HFD on energy homeostasis, inflammation, and hypothalamic and liver gene expression and that restoration of ERE-independent ERα signaling partially reestablishes susceptibility to maternal HFD.
Collapse
Affiliation(s)
- Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA. .,New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Alejandra Villalobos
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Elizabeth A Krumm
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Department of Reproductive Medicine, University of California, San Diego, San Diego, CA 92103, USA
| | - Kyle J Mamounis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.,Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
21
|
Yang JA, Stires H, Belden WJ, Roepke TA. The Arcuate Estrogen-Regulated Transcriptome: Estrogen Response Element-Dependent and -Independent Signaling of ERα in Female Mice. Endocrinology 2017; 158:612-626. [PMID: 28359086 PMCID: PMC5460777 DOI: 10.1210/en.2016-1663] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 12/29/2016] [Indexed: 01/14/2023]
Abstract
To influence energy homeostasis and reproduction, 17β-estradiol (E2) controls the arcuate nucleus (ARC) through multiple receptor-mediated mechanisms, but primarily via estrogen receptor (ER) α, which signals through both estrogen response element (ERE)-dependent and -independent mechanisms. To determine ERα-mediated, ERE-dependent, and ERE-independent E2 signaling in the ARC, we examined the differential regulation of the mouse arcuate transcriptome by E2 using three mice genotypes: (1) wild-type, (2) ERα knock-in/knockout (ERE-independent mechanisms), and (3) total ERα knockout (ERα-independent mechanisms). Females were ovariectomized and injected with oil or E2, and RNA sequencing on the ARC was used to identify E2-regulated genes in each genotype. Our results show that E2 regulates numerous genes involved in cell signaling, cytoskeleton structure, inflammation, neurotransmission, neuropeptide production, and transcription. Furthermore, ERE-independent signaling regulates ARC genes expressed in kisspeptin neurons and transcription factors that control the hypothalamic/pituitary/gonadal axis. Interestingly, a few genes involved in mitochondrial oxidative respiration were regulated by E2 through ERα-independent signaling. A comparison within oil- and E2-treated females across the three genotypes suggests that genes involved in cell growth and proliferation, extracellular matrices, neuropeptides, receptors, and transcription are differentially expressed across the genotypes. Comparing with previously published chromatin immunoprecipitation sequencing analysis, we found that ERE-independent regulation in the ARC is mainly mediated by tethering of ERα, which is consistent with previous findings. We conclude that the mouse arcuate estrogen-regulated transcriptome is regulated by multiple receptor-mediated mechanisms to modulate the central control of energy homeostasis and reproduction, including novel E2-responsive pathways.
Collapse
Affiliation(s)
- Jennifer A Yang
- Department of Animal Sciences and Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Hillary Stires
- Department of Animal Sciences and Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - William J Belden
- Department of Animal Sciences and Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Troy A Roepke
- Department of Animal Sciences and Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
22
|
Yasrebi A, Rivera JA, Krumm EA, Yang JA, Roepke TA. Activation of Estrogen Response Element-Independent ERα Signaling Protects Female Mice From Diet-Induced Obesity. Endocrinology 2017; 158:319-334. [PMID: 27901601 PMCID: PMC5413076 DOI: 10.1210/en.2016-1535] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/29/2016] [Indexed: 01/22/2023]
Abstract
17β-estradiol (E2) regulates central and peripheral mechanisms that control energy and glucose homeostasis predominantly through estrogen receptor α (ERα) acting via receptor binding to estrogen response elements (EREs). ERα signaling is also involved in mediating the effects of E2 on diet-induced obesity (DIO), although the roles of ERE-dependent and -independent ERα signaling in reducing the effects of DIO remain largely unknown. We hypothesize that ERE-dependent ERα signaling is necessary to ameliorate the effects of DIO. We addressed this question using ERα knockout (KO) and ERα knockin/knockout (KIKO) female mice, the latter expressing an ERα that lacks a functional ERE binding domain. Female mice were ovariectomized, fed a low-fat diet (LFD) or a high-fat diet (HFD), and orally dosed with vehicle or estradiol benzoate (EB) (300 μg/kg). After 9 weeks, body composition, glucose and insulin tolerance, peptide hormone and inflammatory cytokine levels, and hypothalamic arcuate nucleus and liver gene expression were assessed. EB reduced body weight and body fat in wild-type (WT) female mice, regardless of diet, and in HFD-fed KIKO female mice, in part by reducing energy intake and feeding efficiency. EB reduced fasting glucose levels in KIKO mice fed both diets but augmented glucose tolerance only in HFD-fed KIKO female mice. Plasma insulin and interleukin 6 were elevated in KIKO and KO female mice compared with LFD-fed WT female mice. Expression of arcuate neuropeptide and receptor genes and liver fatty acid biosynthesis genes was altered by HFD and by EB through ERE-dependent and -independent mechanisms. Therefore, ERE-independent signaling mechanisms in both the brain and peripheral organs mediate, in part, the effects of E2 during DIO.
Collapse
Affiliation(s)
- Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences,
- Graduate Program in Endocrinology and Animal Biosciences, and
| | - Janelle A. Rivera
- Department of Animal Sciences, School of Environmental and Biological Sciences,
| | - Elizabeth A. Krumm
- Department of Animal Sciences, School of Environmental and Biological Sciences,
- Graduate Program in Endocrinology and Animal Biosciences, and
| | - Jennifer A. Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences,
- Graduate Program in Endocrinology and Animal Biosciences, and
| | - Troy A. Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences,
- Graduate Program in Endocrinology and Animal Biosciences, and
- New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08901
| |
Collapse
|
23
|
Yang JA, Yasrebi A, Snyder M, Roepke TA. The interaction of fasting, caloric restriction, and diet-induced obesity with 17β-estradiol on the expression of KNDy neuropeptides and their receptors in the female mouse. Mol Cell Endocrinol 2016; 437:35-50. [PMID: 27507595 PMCID: PMC5048571 DOI: 10.1016/j.mce.2016.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/04/2016] [Accepted: 08/04/2016] [Indexed: 11/27/2022]
Abstract
Arcuate neurons that coexpress kisspeptin (Kiss1), neurokinin B (Tac2), and dynorphin (Pdyn) mediate negative feedback of 17β-estradiol (E2) on the HPG axis. Previous studies report that fasting and caloric restriction reduce arcuate Kiss1 expression. The objective of this study was to determine the interactions of E2 with fasting, caloric restriction, and diet-induced obesity on KNDy gene and receptor expression. Ovariectomized female mice were separated into control and estradiol benzoate (E2B)-treated groups. E2B decreased Kiss1 and the tachykinin 2 receptor, Tac3r, in ARC tissue and Tac2 in Tac2 neurons. Diet-induced obesity decreased Kiss1 in oil-treated animals and the kisspeptin receptor, Kiss1r and Tac3r in the ARC of E2B-treated animals. Chronic caloric (30%) restriction reduced all three neuropeptides in oil-treated females and Kiss1r by E2B in CR animals. Taken together, our experiments suggest that steroidal environment and energy state negatively regulate KNDy gene expression in both ARC and Tac2 neurons.
Collapse
Affiliation(s)
- Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States; Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Marisa Snyder
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States; Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States; Graduate Program in Nutritional Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States; New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, United States.
| |
Collapse
|
24
|
Tran TKA, MacFarlane GR, Kong RYC, O'Connor WA, Yu RMK. Potential mechanisms underlying estrogen-induced expression of the molluscan estrogen receptor (ER) gene. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 179:82-94. [PMID: 27592181 DOI: 10.1016/j.aquatox.2016.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/23/2016] [Indexed: 06/06/2023]
Abstract
In vertebrates, estrogens and estrogen mimicking chemicals modulate gene expression mainly through a genomic pathway mediated by the estrogen receptors (ERs). Although the existence of an ER orthologue in the mollusc genome has been known for some time, its role in estrogen signalling has yet to be deciphered. This is largely due to its constitutive (ligand-independent) activation and a limited mechanistic understanding of its regulation. To fill this knowledge gap, we cloned and characterised an ER cDNA (sgER) and the 5'-flanking region of the gene from the Sydney rock oyster Saccostrea glomerata. The sgER cDNA is predicted to encode a 477-amino acid protein that contains a DNA-binding domain (DBD) and a ligand-binding domain (LBD) typically conserved among both vertebrate and invertebrate ERs. A comparison of the sgER LBD sequence with those of other ligand-dependent ERs revealed that the sgER LBD is variable at several conserved residues known to be critical for ligand binding and receptor activation. Ligand binding assays using fluorescent-labelled E2 and purified sgER protein confirmed that sgER is devoid of estrogen binding. In silico analysis of the sgER 5'-flanking sequence indicated the presence of three putative estrogen responsive element (ERE) half-sites and several putative sites for ER-interacting transcription factors, suggesting that the sgER promoter may be autoregulated by its own gene product. sgER mRNA is ubiquitously expressed in adult oyster tissues, with the highest expression found in the ovary. Ovarian expression of sgER mRNA was significantly upregulated following in vitro and in vivo exposure to 17β-estradiol (E2). Notably, the activation of sgER expression by E2 in vitro was abolished by the specific ER antagonist ICI 182, 780. To determine whether sgER expression is epigenetically regulated, the in vivo DNA methylation status of the putative proximal promoter in ovarian tissues was assessed using bisulfite genomic sequencing. The results showed that the promoter is predominantly hypomethylated (with 0-3.3% methylcytosines) regardless of sgER mRNA levels. Overall, our investigations suggest that the estrogen responsiveness of sgER is regulated by a novel ligand-dependent receptor, presumably via a non-genomic pathway(s) of estrogen signalling.
Collapse
Affiliation(s)
- Thi Kim Anh Tran
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia; Department of Agriculture, Forestry and Fisheries, Vinh University, 182 Le Duan St., Vinh City, Nghe An, Vietnam
| | - Geoff R MacFarlane
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia
| | - Richard Yuen Chong Kong
- Department of Biology and Chemistry, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong Special Administrative Region, China
| | - Wayne A O'Connor
- New South Wales Department of Primary Industries, Port Stephens Fisheries Institute, Taylors Beach, NSW 2316, Australia
| | - Richard Man Kit Yu
- School of Environmental and Life Sciences, The University of Newcastle, Callaghan, NSW 2308, Australia.
| |
Collapse
|
25
|
Hu P, Liu J, Yasrebi A, Gotthardt JD, Bello NT, Pang ZP, Roepke TA. Gq Protein-Coupled Membrane-Initiated Estrogen Signaling Rapidly Excites Corticotropin-Releasing Hormone Neurons in the Hypothalamic Paraventricular Nucleus in Female Mice. Endocrinology 2016; 157:3604-20. [PMID: 27387482 PMCID: PMC5007888 DOI: 10.1210/en.2016-1191] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/01/2016] [Indexed: 02/07/2023]
Abstract
CRH neurons in the hypothalamic paraventricular nucleus (PVN) play a central role in regulating the hypothalamus-pituitary-adrenal (HPA) axis and are directly influenced by 17β-estradiol (E2). Although compelling evidence has suggested the existence of membrane-associated estrogen receptors (mERs) in hypothalamic and other central nervous system neurons, it remains unknown whether E2 impacts CRH neuronal excitability through this mechanism. The purpose of the current study is to examine the existence and function of mER signaling in PVN CRH neurons. Whole-cell recordings were made from CRH neurons identified by Alexa Fluor 594 labeling and post hoc immunostaining in ovariectomized female mice. E2 (100nM) rapidly suppressed the M-current (a voltage-dependent K(+) current) and potentiated glutamatergic excitatory postsynaptic currents. The putative Gq-coupled mER (Gq-mER) characterized in hypothalamic proopiomelanocortin neurons initiates a phospholipase C-protein kinase C-protein kinase A pathway; therefore, we examined the involvement of this pathway using selective inhibitors. Indeed, the ER antagonist ICI 182780 and inhibitors of Gq-phospholipase C-protein kinase C-protein kinase A blocked E2's actions, suggesting dependence on the Gq-mER. Furthermore, STX, a selective ligand for the Gq-mER, mimicked E2's actions. Finally, to examine the in vivo effect of Gq-mER activation, E2 or STX injection increased c-fos expression in CRH neurons in the PVN, suggesting CRH neuronal activation. This corresponded to an increase in plasma corticosterone. We conclude that the Gq-mER plays a critical role in the rapid regulation of CRH neuronal activity and the HPA axis. Our findings provide a potential underlying mechanism for E2's involvement in the pathophysiology of HPA-associated mood disorders.
Collapse
Affiliation(s)
- Pu Hu
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Ji Liu
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Ali Yasrebi
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Juliet D Gotthardt
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Nicholas T Bello
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Zhiping P Pang
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Troy A Roepke
- Department of Animal Sciences (P.H., A.Y., J.D.G., N.T.B., T.A.R.), School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, and Child Health Institute of New Jersey (J.L., Z.P.P.) and Department of Neuroscience and Cell Biology (J.L., Z.P.P.), Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| |
Collapse
|
26
|
Regulation of arcuate genes by developmental exposures to endocrine-disrupting compounds in female rats. Reprod Toxicol 2016; 62:18-26. [PMID: 27103539 DOI: 10.1016/j.reprotox.2016.04.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 01/11/2016] [Accepted: 04/15/2016] [Indexed: 01/09/2023]
Abstract
Developmental exposure to endocrine-disrupting compounds (EDCs) alters reproduction and energy homeostasis, both of which are regulated by the arcuate nucleus (ARC). Little is known about the effects of EDC on ARC gene expression. In Experiment #1, pregnant dams were treated with either two doses of bisphenol A (BPA) or oil from embryonic day (E)18-21. Neonates were injected from postnatal day (PND)0-7. Vaginal opening, body weights, and ARC gene expression were measured. Chrm3 (muscarinic receptor 3) and Adipor1 (adiponectin receptor 1) were decreased by BPA. Bdnf (brain-derived neurotropic factor), Igf1 (insulin-like growth factor 1), Htr2c (5-hydroxytryptamine receptor), and Cck2r (cholescystokinin 2 receptor) were impacted. In Experiment #2, females were exposed to BPA, diethylstilbestrol (DES), di(2-ethylhexyl)phthalate, or methoxychlor (MXC) during E11-PND7. MXC and DES advanced the age of vaginal opening and ARC gene expression was impacted. These data indicate that EDCs alter ARC genes involved in reproduction and energy homeostasis in females.
Collapse
|
27
|
Yang JA, Mamounis KJ, Yasrebi A, Roepke TA. Regulation of gene expression by 17β-estradiol in the arcuate nucleus of the mouse through ERE-dependent and ERE-independent mechanisms. Steroids 2016; 107:128-38. [PMID: 26768413 PMCID: PMC4775315 DOI: 10.1016/j.steroids.2016.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 12/29/2015] [Accepted: 01/03/2016] [Indexed: 12/15/2022]
Abstract
17β-Estradiol (E2) modulates gene expression in the hypothalamic arcuate nucleus (ARC) to control homeostatic functions. In the ARC, estrogen receptor (ER) α is highly expressed and is an important contributor to E2's actions, controlling gene expression through estrogen response element (ERE)-dependent and -independent mechanisms. The objective of this study was to determine if known E2-regulated genes are regulated through these mechanisms. The selected genes have been shown to regulate homeostasis and have been separated into three subsections: channels, receptors, and neuropeptides. To determine if ERE-dependent or ERE-independent mechanisms regulate gene expression, two transgenic mouse models, an ERα knock-out (ERKO) and an ERα knock-in/knock-out (KIKO), which lacks a functional ERE binding domain, were used in addition to their wild-type littermates. Females of all genotypes were ovariectomized and injected with oil or estradiol benzoate (E2B). Our results suggest that E2B regulates multiple genes through these mechanisms. Of note, Cacna1g and Kcnmb1 channel expression was increased by E2B in WT females only, suggesting an ERE-dependent regulation. Furthermore, the NKB receptor, Tac3r, was suppressed by E2B in WT and KIKO females but not ERKO females, suggesting that ERα-dependent, ERE-independent signaling is necessary for Tac3r regulation. The adrenergic receptor Adra1b was suppressed by E2B in all genotypes indicating that ERα is not the primary receptor for E2B's actions. The neuropeptide Tac2 was suppressed by E2B through ERE-dependent mechanisms. These results indicate that E2B activates both ERα-dependent and independent signaling in the ARC through ERE-dependent and ERE-independent mechanisms to control gene expression.
Collapse
Affiliation(s)
- Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Kyle J Mamounis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
28
|
Yasrebi A, Hsieh A, Mamounis KJ, Krumm EA, Yang JA, Magby J, Hu P, Roepke TA. Differential gene regulation of GHSR signaling pathway in the arcuate nucleus and NPY neurons by fasting, diet-induced obesity, and 17β-estradiol. Mol Cell Endocrinol 2016; 422:42-56. [PMID: 26577678 PMCID: PMC4742417 DOI: 10.1016/j.mce.2015.11.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 12/20/2022]
Abstract
Ghrelin's receptor, growth hormone secretagogue receptor (GHSR), is highly expressed in the arcuate nucleus (ARC) and in neuropeptide Y (NPY) neurons. Fasting, diet-induced obesity (DIO), and 17β-estradiol (E2) influence ARC Ghsr expression. It is unknown if these effects occur in NPY neurons. Therefore, we examined the expression of Npy, Agrp, and GHSR signaling pathway genes after fasting, DIO, and E2 replacement in ARC and pools of NPY neurons. In males, fasting increased ARC Ghsr and NPY Foxo1 but decreased NPY Ucp2. In males, DIO decreased ARC and NPY Ghsr and Cpt1c. In fed females, E2 increased Agrp, Ghsr, Cpt1c, and Foxo1 in ARC. In NPY pools, E2 decreased Foxo1 in fed females but increased Foxo1 in fasted females. DIO in females suppressed Agrp and augmented Cpt1c in NPY neurons. In summary, genes involved in GHSR signaling are differentially regulated between the ARC and NPY neurons in a sex-dependent manner.
Collapse
Affiliation(s)
- Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Anna Hsieh
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Kyle J Mamounis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Elizabeth A Krumm
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Jason Magby
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Pu Hu
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; Graduate Program in Endocrinology and Animal Biosciences, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA; New Jersey Institute for Food, Nutrition, and Health, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
29
|
Wagner EJ. Sex differences in cannabinoid-regulated biology: A focus on energy homeostasis. Front Neuroendocrinol 2016; 40:101-9. [PMID: 26800649 PMCID: PMC4783283 DOI: 10.1016/j.yfrne.2016.01.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 12/31/2015] [Accepted: 01/19/2016] [Indexed: 11/18/2022]
Abstract
Considerable strides have been made over the past 20 years in our understanding of the ligands, receptor subtypes, signal transduction mechanisms and biological actions comprising the endocannabinoid system. From the ever-expanding number of studies that have been conducted during this time, it has become increasingly clear that sex differences are the cornerstone of cannabinoid-regulated biology. Available evidence has demonstrated that these sex differences endure in the absence of gonadal steroids, and are modulated by the acute, activational effects of these hormones. This review focuses on select aspects of sexually differentiated, cannabinoid-regulated biology, with a particular emphasis on the control of energy balance. It is anticipated that it will lend impactful insight into the pervasive and diverse disparities in how males and females respond to cannabinoids--from the organismal level down to the molecular level. Additionally, it will furnish a newfound appreciation for the need to recalibrate our thinking in terms of how cannabinoids are used as therapeutic adjuvants for a broad range of clinical disorders and associated comorbidities, including body wasting and obesity.
Collapse
Affiliation(s)
- Edward J Wagner
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766, United States.
| |
Collapse
|
30
|
Tang YR, Yang WW, Wang Y, Gong YY, Jiang LQ, Lin L. Estrogen regulates the expression of small-conductance Ca-activated K+ channels in colonic smooth muscle cells. Digestion 2015; 91:187-96. [PMID: 25790748 DOI: 10.1159/000371544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/12/2014] [Indexed: 02/04/2023]
Abstract
AIM This study aimed to determine the effects of small-conductance Ca(2+)-activated K(+) (SK) channels in colonic relaxation and the regulation of SK channels by estrogen. METHODS The contractile activity of muscle strips from male rats was estimated, and drugs including vehicle (DMSO), 17β-estradiol (E2), or apamin (SK blocker) were added, respectively. In a further experiment, muscle strips were preincubated with apamin before exposure to E2. The levels of the SK2 and SK3 protein expression in the colonic smooth muscle cells (SMCs) were detected. SMCs were treated with ICI 182780 (estrogen receptor [ER] antagonist) plus E2, BSA-E2, PPT (ERα agonist), or DPN (ERβ agonist). SK3 mRNA and protein expression levels were detected. RESULTS The muscle strips responded to E2 with a decrease and to apamin with a transient increase in tension. Preincubation with apamin partially prevented E2-induced relaxation. Two SK channel subtypes, SK2 and SK3, were coexpressed with α-actin in colonic SMCs. The quantitative ratio of the SK transcriptional expression in colonic SMCs was SK3 > SK2. The SK3 expression was upregulated by E2, and was downregulated by ICI 182780, but was not influenced by BSA-E2. Furthermore, the effect of PPT on the expression of SK3 was almost the same as that of E2, while DPN did not influence the protein expression of SK3. CONCLUSION These findings indicate that SK3 is involved in the E2-induced relaxing effect on rat colonic smooth muscle. Furthermore, E2 upregulates the expression of SK3 in rat SMCs, and that this effect is mediated via the ERα receptor.
Collapse
Affiliation(s)
- Yu-Rong Tang
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
31
|
Kelly MJ, Rønnekleiv OK. Minireview: neural signaling of estradiol in the hypothalamus. Mol Endocrinol 2015; 29:645-57. [PMID: 25751314 PMCID: PMC4415204 DOI: 10.1210/me.2014-1397] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Affiliation(s)
- Martin J Kelly
- Departments of Physiology and Pharmacology (M.J.K., O.K.R.) and Anesthesiology and Perioperative Medicine (O.K.R.), Oregon Health and Science University, Portland, Oregon 97239; and Division of Neuroscience (M.J.K., O.K.R.), Oregon National Primate Research Center; Oregon Health and Science University, Beaverton, Oregon 97006
| | | |
Collapse
|
32
|
Wellhauser L, Gojska NM, Belsham DD. Delineating the regulation of energy homeostasis using hypothalamic cell models. Front Neuroendocrinol 2015; 36:130-49. [PMID: 25223866 DOI: 10.1016/j.yfrne.2014.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/28/2014] [Accepted: 09/02/2014] [Indexed: 12/27/2022]
Abstract
Attesting to its intimate peripheral connections, hypothalamic neurons integrate nutritional and hormonal cues to effectively manage energy homeostasis according to the overall status of the system. Extensive progress in the identification of essential transcriptional and post-translational mechanisms regulating the controlled expression and actions of hypothalamic neuropeptides has been identified through the use of animal and cell models. This review will introduce the basic techniques of hypothalamic investigation both in vivo and in vitro and will briefly highlight the key advantages and challenges of their use. Further emphasis will be place on the use of immortalized models of hypothalamic neurons for in vitro study of feeding regulation, with a particular focus on cell lines proving themselves most fruitful in deciphering fundamental basics of NPY/AgRP, Proglucagon, and POMC neuropeptide function.
Collapse
Affiliation(s)
- Leigh Wellhauser
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Nicole M Gojska
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Denise D Belsham
- Departments of Physiology, Medicine and OB/GYN, University of Toronto, Toronto, Ontario M5G 1A8, Canada; Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
33
|
Fontana R, Della Torre S, Meda C, Longo A, Eva C, Maggi AC. Estrogen replacement therapy regulation of energy metabolism in female mouse hypothalamus. Endocrinology 2014; 155:2213-21. [PMID: 24635349 DOI: 10.1210/en.2013-1731] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Estrogens play an important role in the regulation of energy homeostasis in female mammals and a reduced ovarian function, due to natural aging or surgery, is associated with body weight increase and fat redistribution. This disruption of energy homeostasis may constitute a trigger for several pathologies known to be associated with climacterium; however, so far, limited attention has been devoted to the ability of estrogen replacement therapies (ERT) to reinstate the balanced energy metabolism characteristic of cycling female mammals. The purpose of the present study was to compare the efficacy of selected ERTs in reversing the ovariectomy-induced gain in body weight. To this aim female ERE-Luc mice were ovariectomized and, after 3 weeks, treated per os for 21 days with: conjugated estrogens, two selective estrogen receptor modulators (bazedoxifene and raloxifene), and the combination of bazedoxifene plus conjugated estrogens (tissue-selective estrogen complex, TSEC). The study shows that the therapy based on TSEC was the most efficacious in reducing the body weight accrued by ovariectomy (OVX). In addition, by means of in vivo imaging, the TSEC treatment was shown to increase estrogen receptor (ER) transcriptional activity selectively in the arcuate nucleus, which is a key area for the control of energy homeostasis. Finally, quantitative analysis of the mRNAs encoding orexigenic and anorexigenic peptides indicated that following ERT with TSEC there was a significant change in Agrp, NPY, and Kiss-1 mRNA accumulation in the whole hypothalamus. Considering that prior studies showed that ERT with TSEC was able to mimic the rhythm of ER oscillatory activity during the reproductive cycle and that such fluctuations were relevant for energy metabolism, the present observations further point to the ER tetradian oscillation as an important component of the ER signaling necessary for the full hormone action and therefore for an efficacious ERT.
Collapse
Affiliation(s)
- Roberta Fontana
- Center of Excellence on Neurodegenerative Diseases and Department of Pharmacological and Biomolecular Sciences (R.F., S.D.T, C.M., A.M), University of Milan, 21022 Milan, Italy; Department of Drug Discovery and Development, Italian Institute of Technology, 16163 Genova, Italy (R.F.); Neuroscience Institute Cavalieri Ottolenghi, (A.L., E.C.) 10043 Turin, Italy
| | | | | | | | | | | |
Collapse
|
34
|
Nestor CC, Kelly MJ, Rønnekleiv OK. Cross-talk between reproduction and energy homeostasis: central impact of estrogens, leptin and kisspeptin signaling. Horm Mol Biol Clin Investig 2014; 17:109-28. [PMID: 25372735 PMCID: PMC4959432 DOI: 10.1515/hmbci-2013-0050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/07/2013] [Indexed: 02/06/2023]
Abstract
The central nervous system receives hormonal cues (e.g., estrogens and leptin, among others) that influence reproduction and energy homeostasis. 17β-estradiol (E2) is known to regulate gonadotropin-releasing hormone (GnRH) secretion via classical steroid signaling and rapid non-classical membrane-initiated signaling. Because GnRH neurons are void of leptin receptors, the actions of leptin on these neurons must be indirect. Although it is clear that the arcuate nucleus of the hypothalamus is the primary site of overlap between these two systems, it is still unclear which neural network(s) participate in the cross-talk of E2 and leptin, two hormones essential for reproductive function and metabolism. Herein we review the progress made in understanding the interactions between reproduction and energy homeostasis by focusing on the advances made to understand the cellular signaling of E2 and leptin on three neural networks: kisspeptin, pro-opiomelanocortin (POMC) and neuropeptide Y (NPY). Although critical in mediating the actions of E2 and leptin, considerable work still remains to uncover how these neural networks interact in vivo.
Collapse
Affiliation(s)
- Casey C Nestor
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA; and Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA; and Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| |
Collapse
|
35
|
Mamounis KJ, Yang JA, Yasrebi A, Roepke TA. Estrogen response element-independent signaling partially restores post-ovariectomy body weight gain but is not sufficient for 17β-estradiol's control of energy homeostasis. Steroids 2014; 81:88-98. [PMID: 24252383 PMCID: PMC3947695 DOI: 10.1016/j.steroids.2013.10.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The steroid 17β-estradiol (E2) modulates energy homeostasis by reducing feeding behavior and increasing energy expenditure primarily through estrogen receptor α (ERα)-mediated mechanisms. Intact ERαKO female mice develop obesity as adults exhibiting decreased energy expenditure and increased fat deposition. However, intact transgenic female mice expressing a DNA-binding-deficient ERα (KIKO) are not obese and have similar energy expenditure, activity and fat deposition as to wild type (WT) females, suggesting that non-estrogen response element (ERE)-mediated signaling is important in E2 regulation of energy homeostasis. Initial reports did not examine the effects of ovariectomy on energy homeostasis or E2's attenuation of post-ovariectomy body weight gain. Therefore, we sought to determine if low physiological doses of E2 (250 ng QOD) known to suppress post-ovariectomy body weight gain in WT females would suppress body weight gain in ovariectomized KIKO females. We observed that the post-ovariectomy increase in body weight was significantly greater in WT females than in KIKO females. Furthermore, E2 did not significantly attenuate the body weight gain in KIKO females as it did in WT females. E2 replacement suppressed food intake and fat accumulation while increasing nighttime oxygen consumption and activity only in WT females. E2 replacement also increased arcuate POMC gene expression in WT females only. These data suggest that in the intact female, ERE-independent mechanisms are sufficient to maintain normal energy homeostasis and to partially restore the normal response to ovariectomy. However, they are not sufficient for E2's suppression of post-ovariectomy body weight gain and its effects on metabolism and activity.
Collapse
Affiliation(s)
- Kyle J Mamounis
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jennifer A Yang
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Ali Yasrebi
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, USA
| | - Troy A Roepke
- Department of Animal Sciences, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, USA.
| |
Collapse
|
36
|
Abstract
Two populations of cells within the hypothalamus exert opposite actions on food intake: proopiomelanocortin (POMC) neurons decrease it, while neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons increase it. 17β-Estradiol (E2) is a potent anorexigenic hormone that exerts both genomic and non-genomic, rapid actions on these metabolic neurons. This review focuses on the rapid membrane effects of E2 in both POMC and NPY/AgRP neurons and how these combined effects mediate the anorexigenic effects of this steroid.
Collapse
Affiliation(s)
- A W Smith
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA
| | - O K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - M J Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR 97239, USA; Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| |
Collapse
|
37
|
Smith AW, Bosch MA, Wagner EJ, Rønnekleiv OK, Kelly MJ. The membrane estrogen receptor ligand STX rapidly enhances GABAergic signaling in NPY/AgRP neurons: role in mediating the anorexigenic effects of 17β-estradiol. Am J Physiol Endocrinol Metab 2013; 305:E632-40. [PMID: 23820624 PMCID: PMC3761166 DOI: 10.1152/ajpendo.00281.2013] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Besides its quintessential role in reproduction, 17β-estradiol (E2) is a potent anorexigenic hormone. E2 and the selective Gq-coupled membrane estrogen receptor (Gq-mER) ligand STX rapidly increase membrane excitability in proopiomelanocortin (POMC) neurons by desensitizing the coupling of GABAB receptors to G protein-coupled inwardly rectifying K(+) channels (GIRKs), which upon activation elicit a hyperpolarizing outward current. However, it is unknown whether E2 and STX can modulate GABAB signaling in neuropeptide Y (NPY)/agouti-related peptide (AgRP) neurons. We used single-cell RT-PCR and whole cell patch clamping with selective pharmacological reagents to show that NPY/AgRP cells of mice express the GABAB-R1 and -R2 receptors and are hyperpolarized by the GABAB agonist baclofen in an E2-dependent manner. In males, E2 rapidly attenuated the coupling of GABAB receptors to GIRKs, which was blocked by the general PI3K inhibitors wortmannin and LY-294002 or the selective p110β subunit inhibitor TGX-221. The ERα-selective agonist propyl pyrazole triol mimicked the effects of E2. STX, in contrast, enhanced the GABAB response in males, which was abrogated by the estrogen receptor (ER) antagonist ICI 182,780. In gonadectomized mice of both sexes, E2 enhanced or attenuated the GABAB response in different NPY/AgRP cells. Coperfusing wortmannin with E2 or simply applying STX always enhanced the GABAB response. Thus, in NPY/AgRP neurons, activation of the Gq-mER by E2 or STX enhances the GABAergic postsynaptic response, whereas activation of ERα by E2 attenuates it. These findings demonstrate a clear functional dichotomy of rapid E2 membrane-initiated signaling via ERα vs. Gq-mER in a CNS neuron vital for regulating energy homeostasis.
Collapse
Affiliation(s)
- A W Smith
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | | | | | | | | |
Collapse
|
38
|
Kelly MJ, Rønnekleiv OK. A selective membrane estrogen receptor agonist maintains autonomic functions in hypoestrogenic states. Brain Res 2013; 1514:75-82. [PMID: 23535448 PMCID: PMC5432040 DOI: 10.1016/j.brainres.2013.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 03/09/2013] [Accepted: 03/19/2013] [Indexed: 12/14/2022]
Abstract
It is well known that many of the actions of estrogens in the central nervous system are mediated via intracellular receptor/transcription factors that interact with steroid response elements on target genes. But there is also a compelling evidence for the involvement of membrane estrogen receptors in hypothalamic and other CNS functions. However, it is not well understood how estrogens signal via membrane receptors, and how these signals impact not only membrane excitability but also gene transcription in neurons. Indeed, it has been known for sometime that estrogens can rapidly alter neuronal activity within seconds, indicating that some cellular effects can occur via membrane delimited events. In addition, estrogens can affect second messenger systems including calcium mobilization and a plethora of kinases within neurons to alter cellular functions. Therefore, this brief review will summarize our current understanding of rapid membrane-initiated and intracellular signaling by estrogens in the hypothalamus, the nature of receptors involved and how these receptors contribute to maintenance of homeostatic functions, many of which go awry in menopausal states. This article is part of a Special Issue entitled Hormone Therapy.
Collapse
Affiliation(s)
- Martin J Kelly
- Department of Physiology and Pharmacology, L334, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239-3098, USA.
| | | |
Collapse
|
39
|
|
40
|
Shi H, Kumar SPDS, Liu X. G protein-coupled estrogen receptor in energy homeostasis and obesity pathogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 114:193-250. [PMID: 23317786 PMCID: PMC3632385 DOI: 10.1016/b978-0-12-386933-3.00006-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity and its related metabolic diseases have reached a pandemic level worldwide. There are sex differences in the prevalence of obesity and its related metabolic diseases, with men being more vulnerable than women; however, the prevalence of these disorders increases dramatically in women after menopause, suggesting that sex steroid hormone estrogens play key protective roles against development of obesity and metabolic diseases. Estrogens are important regulators of several aspects of metabolism, including body weight and body fat, caloric intake and energy expenditure, and glucose and lipid metabolism in both males and females. Estrogens act in complex ways on their nuclear estrogen receptors (ERs) ERα and ERβ and transmembrane ERs such as G protein-coupled estrogen receptor. Genetic tools, such as different lines of knockout mouse models, and pharmacological agents, such as selective agonists and antagonists, are available to study function and signaling mechanisms of ERs. We provide an overview of the evidence for the physiological and cellular actions of ERs in estrogen-dependent processes in the context of energy homeostasis and body fat regulation and discuss its pathology that leads to obesity and related metabolic states.
Collapse
Affiliation(s)
- Haifei Shi
- Department of Biology, Center for Physiology and Neuroscience, Miami University, Oxford, Ohio, USA
| | | | | |
Collapse
|
41
|
Kelly MJ, Rønnekleiv OK. Membrane-initiated actions of estradiol that regulate reproduction, energy balance and body temperature. Front Neuroendocrinol 2012; 33:376-87. [PMID: 22871514 PMCID: PMC3618441 DOI: 10.1016/j.yfrne.2012.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 07/07/2012] [Accepted: 07/11/2012] [Indexed: 12/21/2022]
Abstract
It is well known that many of the actions of estrogens in the central nervous system are mediated via intracellular receptor/transcription factors that interact with steroid response elements on target genes. However, there now exists compelling evidence for membrane estrogen receptors in hypothalamic and other brain neurons. But, it is not well understood how estrogens signal via membrane receptors, and how these signals impact not only membrane excitability but also gene transcription in neurons. Indeed, it has been known for sometime that estrogens can rapidly alter neuronal activity within seconds, indicating that some cellular effects can occur via membrane delimited events. In addition, estrogens can affect second messenger systems including calcium mobilization and a plethora of kinases to alter cell signaling. Therefore, this review will consider our current knowledge of rapid membrane-initiated and intracellular signaling by estrogens in the hypothalamus, the nature of receptors involved and how they contribute to homeostatic functions.
Collapse
Affiliation(s)
- Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
- Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
| | - Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239
- Division of Neuroscience, Oregon Regional Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
42
|
Sinchak K, Wagner EJ. Estradiol signaling in the regulation of reproduction and energy balance. Front Neuroendocrinol 2012; 33:342-63. [PMID: 22981653 PMCID: PMC3496056 DOI: 10.1016/j.yfrne.2012.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Our knowledge of membrane estrogenic signaling mechanisms and their interactions that regulate physiology and behavior has grown rapidly over the past three decades. The discovery of novel membrane estrogen receptors and their signaling mechanisms has started to reveal the complex timing and interactions of these various signaling mechanisms with classical genomic steroid actions within the nervous system to regulate physiology and behavior. The activation of the various estrogenic signaling mechanisms is site specific and differs across the estrous cycle acting through both classical genomic mechanisms and rapid membrane-initiated signaling to coordinate reproductive behavior and physiology. This review focuses on our current understanding of estrogenic signaling mechanisms to promote: (1) sexual receptivity within the arcuate nucleus of the hypothalamus, (2) estrogen positive feedback that stimulates de novo neuroprogesterone synthesis to trigger the luteinizing hormone surge important for ovulation and estrous cyclicity, and (3) alterations in energy balance.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Blvd., Long Beach, CA 90840-9502, United States.
| | | |
Collapse
|
43
|
Micevych PE, Kelly MJ. Membrane estrogen receptor regulation of hypothalamic function. Neuroendocrinology 2012; 96:103-10. [PMID: 22538318 PMCID: PMC3496782 DOI: 10.1159/000338400] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 03/18/2012] [Indexed: 11/19/2022]
Abstract
Over the decades, our understanding of estrogen receptor (ER) function has evolved. Today we are confronted by at least two nuclear ERs, ERα and ERβ, and a number of putative membrane ERs, including ERα, ERβ, ER-X, GPR30 and Gq-mER. These receptors all bind estrogens or at least estrogenic compounds and activate intracellular signaling pathways. In some cases, a well-defined pharmacology and physiology has been discovered. In other cases, the identity or the function remains to be elucidated. This mini-review attempts to synthesize our understanding of 17β-estradiol membrane signaling within hypothalamic circuits involved in homeostatic functions, focusing on reproduction and energy balance.
Collapse
Affiliation(s)
- Paul E. Micevych
- Department of Neurobiology, David Geffen School of Medicine, Brain Research Institute, University of California Los Angeles, CA USA
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR USA
| |
Collapse
|
44
|
Washburn N, Borgquist A, Wang K, Jeffery GS, Kelly MJ, Wagner EJ. Receptor subtypes and signal transduction mechanisms contributing to the estrogenic attenuation of cannabinoid-induced changes in energy homeostasis. Neuroendocrinology 2012; 97:160-75. [PMID: 22538462 PMCID: PMC3702272 DOI: 10.1159/000338669] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 04/02/2012] [Indexed: 01/21/2023]
Abstract
We examined the receptor subtypes and signal transduction mechanisms contributing to the estrogenic modulation of cannabinoid-induced changes in energy balance. Food intake and, in some cases, O2 consumption, CO2 production and the respiratory exchange ratio were evaluated in ovariectomized female guinea pigs treated s.c. with the cannabinoid receptor agonist WIN 55,212-2 or its cremephor/ethanol/0.9% saline vehicle, and either with estradiol benzoate (EB), the estrogen receptor (ER) α agonist PPT, the ERβ agonist DPN, the Gq-coupled membrane ER agonist STX, the GPR30 agonist G-1 or their respective vehicles. Patch-clamp recordings were performed in hypothalamic slices. EB, STX, PPT and G-1 decreased daily food intake. Of these, EB, STX and PPT blocked the WIN 55,212-2-induced increase in food intake within 1-4 h. The estrogenic diminution of cannabinoid-induced hyperphagia correlated with a rapid (within 15 min) attenuation of cannabinoid-mediated decreases in glutamatergic synaptic input onto arcuate neurons, which was completely blocked by inhibition of protein kinase C (PKC) and attenuated by inhibition of protein kinase A (PKA). STX, but not PPT, mimicked this rapid estrogenic effect. However, PPT abolished the cannabinoid-induced inhibition of glutamatergic neurotransmission in cells from animals treated 24 h prior. The estrogenic antagonism of this presynaptic inhibition was observed in anorexigenic proopiomelanocortin neurons. These data reveal that estrogens negatively modulate cannabinoid-induced changes in energy balance via Gq-coupled membrane ER- and ERα-mediated mechanisms involving activation of PKC and PKA. As such, they further our understanding of the pathways through which estrogens act to temper cannabinoid sensitivity in regulating energy homeostasis in females.
Collapse
Affiliation(s)
- Neal Washburn
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Amanda Borgquist
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Kate Wang
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Garrett S. Jeffery
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Martin J. Kelly
- Department of Physiology & Pharmacology, Oregon Health & Science University, Portland OR, USA
| | - Edward J. Wagner
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
45
|
Ropero AB, Alonso-Magdalena P, Soriano S, Juan-Picó P, Roepke TA, Kelly MJ, Nadal Á. Insulinotropic effect of the non-steroidal compound STX in pancreatic β-cells. PLoS One 2012; 7:e34650. [PMID: 22506040 PMCID: PMC3323542 DOI: 10.1371/journal.pone.0034650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 03/05/2012] [Indexed: 11/30/2022] Open
Abstract
The non-steroidal compound STX modulates the hypothalamic control of core body temperature and energy homeostasis. The aim of this work was to study the potential effects of STX on pancreatic β-cell function. 1–10 nM STX produced an increase in glucose-induced insulin secretion in isolated islets from male mice, whereas it had no effect in islets from female mice. This insulinotropic effect of STX was abolished by the anti-estrogen ICI 182,780. STX increased intracellular calcium entry in both whole islets and isolated β-cells, and closed the KATP channel, suggesting a direct effect on β-cells. When intraperitoneal glucose tolerance test was performed, a single dose of 100 µg/kg body weight STX improved glucose sensitivity in males, yet it had a slight effect on females. In agreement with the effect on isolated islets, 100 µg/kg dose of STX enhanced the plasma insulin increase in response to a glucose load, while it did not in females. Long-term treatment (100 µg/kg, 6 days) of male mice with STX did not alter body weight, fasting glucose, glucose sensitivity or islet insulin content. Ovariectomized females were insensitive to STX (100 µg/kg), after either an acute administration or a 6-day treatment. This long-term treatment was also ineffective in a mouse model of mild diabetes. Therefore, STX appears to have a gender-specific effect on blood glucose homeostasis, which is only manifested after an acute administration. The insulinotropic effect of STX in pancreatic β-cells is mediated by the closure of the KATP channel and the increase in intracellular calcium concentration. The in vivo improvement in glucose tolerance appears to be mostly due to the enhancement of insulin secretion from β-cells.
Collapse
Affiliation(s)
- Ana B Ropero
- Instituto de Bioingeniería and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Universidad Miguel Hernández, Elche, Spain.
| | | | | | | | | | | | | |
Collapse
|
46
|
Choleris E, Clipperton-Allen AE, Phan A, Valsecchi P, Kavaliers M. Estrogenic involvement in social learning, social recognition and pathogen avoidance. Front Neuroendocrinol 2012; 33:140-59. [PMID: 22369749 DOI: 10.1016/j.yfrne.2012.02.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 02/13/2012] [Accepted: 02/14/2012] [Indexed: 12/25/2022]
Abstract
Sociality comes with specific cognitive skills that allow the proper processing of information about others (social recognition), as well as of information originating from others (social learning). Because sociality and social interactions can also facilitate the spread of infection among individuals the ability to recognize and avoid pathogen threat is also essential. We review here various studies primarily from the rodent literature supporting estrogenic involvement in the regulation of social recognition, social learning (socially acquired food preferences and mate choice copying) and the recognition and avoidance of infected and potentially infected individuals. We consider both genomic and rapid estrogenic effects involving estrogen receptors α and β, and G-protein coupled estrogen receptor 1, along with their interactions with neuropeptide systems in the processing of social stimuli and the regulation and expression of these various socially relevant behaviors.
Collapse
Affiliation(s)
- Elena Choleris
- Department of Psychology, University of Guelph, Guelph, Ontario, Canada N1G 2W1.
| | | | | | | | | |
Collapse
|
47
|
Abstract
17β-Oestradiol (E(2)) is essential for cyclical gonadotrophin-releasing hormone (GnRH) neuronal activity and secretion. In particular, E(2) increases the excitability of GnRH neurones during the afternoon of pro-oestrus in the rodent, which is associated with increased synthesis and secretion of GnRH. It is well established that E(2) regulates the activity of GnRH neurones through both presynaptic and postsynaptic mechanisms. E(2) significantly modulates the mRNA expression of numerous ion channels in GnRH neurones and alters the associated endogenous conductances, including potassium (K(ATP) , A-type) currents and low-voltage T-type and high-voltage L-type calcium currents. Notably, K(ATP) channels are critical for maintaining GnRH neurones in a hyperpolarised state for recruiting the T-type calcium channels, which are important for burst firing in GnRH neurones. In addition, there are other critical channels contributing to burst firing pattern, including the small conductance Ca(2+) -activated K(+) channels that may be modulated by E(2) . Despite these advances, the cellular mechanisms underlying the cyclical GnRH neuronal activity and GnRH release are largely unknown. Ultimately, the ensemble of both pre- and postsynaptic targets of the actions of E(2) will dictate the excitability and activity pattern of GnRH neurones.
Collapse
Affiliation(s)
- O K Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, OR 97239-3098, USA.
| | | | | |
Collapse
|
48
|
Fasting and 17β-estradiol differentially modulate the M-current in neuropeptide Y neurons. J Neurosci 2011; 31:11825-35. [PMID: 21849543 DOI: 10.1523/jneurosci.1395-11.2011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multiple K(+) conductances are targets for many peripheral and central signals involved in the control of energy homeostasis. Potential K(+) channel targets are the KCNQ subunits that form the channels underlying the M-current, a subthreshold, non-inactivating K(+) current that is a common target for G-protein-coupled receptors. Whole-cell recordings were made from GFP (Renilla)-tagged neuropeptide Y (NPY) neurons from the arcuate nucleus of the hypothalamus using protocols to isolate and characterize the M-current in these orexigenic neurons. We recorded robust K(+) currents in the voltage range of the M-current, which were inhibited by the selective KCNQ channel blocker 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone dihydrochloride (XE991) (40 μm), in both intact males and ovariectomized, 17β-estradiol (E2)-treated females. Since NPY neurons are orexigenic and are active during fasting, the M-current was measured in fed and fasted male mice. Fasting attenuated the XE991-sensitive current by threefold, which correlated with decreased expression of the KCNQ2 and KCNQ3 subunits as measured with quantitative real-time PCR. Furthermore, E2 treatment augmented the XE991-sensitive M-current by threefold in ovariectomized (vs oil-treated) female mice. E2 treatment increased the expression of the KCNQ5 subunit in females but not KCNQ2 or KCNQ3 subunits. Fasting in females abrogated the effects of E2 on M-current activity, at least in part, by decreasing KCNQ2 and KCNQ3 expression. In summary, these data suggest that the M-current plays a pivotal role in the modulation of NPY neuronal excitability and may be an important cellular target for neurotransmitter and hormonal signals in the control of energy homeostasis in both males and females.
Collapse
|
49
|
Kenealy BP, Keen KL, Terasawa E. Rapid action of estradiol in primate GnRH neurons: the role of estrogen receptor alpha and estrogen receptor beta. Steroids 2011; 76:861-6. [PMID: 21354432 PMCID: PMC3183999 DOI: 10.1016/j.steroids.2011.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/07/2011] [Accepted: 02/16/2011] [Indexed: 01/08/2023]
Abstract
Estrogens play a pivotal role in the control of female reproductive function. Recent studies using primate GnRH neurons derived from embryonic nasal placode indicate that 17β-estradiol (E(2)) causes a rapid stimulatory action. E(2) (1nM) stimulates firing activity and intracellular calcium ([Ca(2+)](i)) oscillations of primate GnRH neurons within a few min. E(2) also stimulates GnRH release within 10min. However, the classical estrogen receptors, ERα and ERβ, do not appear to play a role in E(2)-induced [Ca(2+)](i) oscillations or GnRH release, as the estrogen receptor antagonist, ICI 182,780, failed to block these responses. Rather, this rapid E(2) action is, at least in part, mediated by a G-protein coupled receptor GPR30. In the present study we further investigate the role of ERα and ERβ in the rapid action of E(2) by knocking down cellular ERα and ERβ by transfection of GnRH neurons with specific siRNA for rhesus monkey ERα and ERβ. Results indicate that cellular knockdown of ERα and ERβ failed to block the E(2)-induced changes in [Ca(2+)](i) oscillations. It is concluded that neither ERα nor ERβ is required for the rapid action of E(2) in primate GnRH neurons.
Collapse
Affiliation(s)
- B P Kenealy
- Wisconsin National Primate Research Center, Madison, WI 53715, USA
| | | | | |
Collapse
|
50
|
Eckel LA. The ovarian hormone estradiol plays a crucial role in the control of food intake in females. Physiol Behav 2011; 104:517-24. [PMID: 21530561 DOI: 10.1016/j.physbeh.2011.04.014] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 04/13/2011] [Accepted: 04/13/2011] [Indexed: 01/05/2023]
Abstract
Despite a strong male bias in both basic and clinical research, it is becoming increasingly accepted that the ovarian hormone estradiol plays an important role in the control of food intake in females. Estradiol's feeding inhibitory effect occurs in a variety of species, including women, but the underlying mechanism has been studied most extensively in rats and mice. Accordingly, much of the data reviewed here is derived from the rodent literature. Adult female rats display a robust decrease in food intake during estrus and ovariectomy promotes hyperphagia and weight gain, both of which can be prevented by a physiological regimen of estradiol treatment. Behavioral analyses have demonstrated that the feeding inhibitory effect of estradiol is mediated entirely by a decrease in meal size. In rats, estradiol appears to exert this action indirectly via interactions with peptide and neurotransmitter systems implicated in the direct control of meal size. Here, I summarize research examining the neurobiological mechanism underlying estradiol's anorexigenic effect. Central estrogen receptors (ERs) have been implicated and activation of one ER subtype in particular, ERα, appears both sufficient and necessary for the estrogenic control of food intake. Future studies are necessary to identify the critical brain areas and intracellular signaling pathways responsible for estradiol's anorexigenic effect. A clearer understanding of the estrogenic control of food intake is prerequisite to elucidating the biological factors that contribute to obesity and eating disorders, both of which are more prevalent in women, compared to men.
Collapse
Affiliation(s)
- Lisa A Eckel
- Program in Neuroscience, Florida State University, 1107 West Call Street,Tallahassee, FL 32306-4301, USA.
| |
Collapse
|