1
|
Anger JT, Case LK, Baranowski AP, Berger A, Craft RM, Damitz LA, Gabriel R, Harrison T, Kaptein K, Lee S, Murphy AZ, Said E, Smith SA, Thomas DA, Valdés Hernández MDC, Trasvina V, Wesselmann U, Yaksh TL. Pain mechanisms in the transgender individual: a review. FRONTIERS IN PAIN RESEARCH 2024; 5:1241015. [PMID: 38601924 PMCID: PMC11004280 DOI: 10.3389/fpain.2024.1241015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 01/25/2024] [Indexed: 04/12/2024] Open
Abstract
Specific Aim Provide an overview of the literature addressing major areas pertinent to pain in transgender persons and to identify areas of primary relevance for future research. Methods A team of scholars that have previously published on different areas of related research met periodically though zoom conferencing between April 2021 and February 2023 to discuss relevant literature with the goal of providing an overview on the incidence, phenotype, and mechanisms of pain in transgender patients. Review sections were written after gathering information from systematic literature searches of published or publicly available electronic literature to be compiled for publication as part of a topical series on gender and pain in the Frontiers in Pain Research. Results While transgender individuals represent a significant and increasingly visible component of the population, many researchers and clinicians are not well informed about the diversity in gender identity, physiology, hormonal status, and gender-affirming medical procedures utilized by transgender and other gender diverse patients. Transgender and cisgender people present with many of the same medical concerns, but research and treatment of these medical needs must reflect an appreciation of how differences in sex, gender, gender-affirming medical procedures, and minoritized status impact pain. Conclusions While significant advances have occurred in our appreciation of pain, the review indicates the need to support more targeted research on treatment and prevention of pain in transgender individuals. This is particularly relevant both for gender-affirming medical interventions and related medical care. Of particular importance is the need for large long-term follow-up studies to ascertain best practices for such procedures. A multi-disciplinary approach with personalized interventions is of particular importance to move forward.
Collapse
Affiliation(s)
- Jennifer T. Anger
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Laura K. Case
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| | - Andrew P. Baranowski
- Pelvic Pain Medicine and Neuromodulation, University College Hospital Foundation Trust, University College London, London, United Kingdom
| | - Ardin Berger
- Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Rebecca M. Craft
- Department of Psychology, Washington State University, Pullman, WA, United States
| | - Lyn Ann Damitz
- Division of Plastic and Reconstructive Surgery, University of North Carolina, Chapel Hill, NC, United States
| | - Rodney Gabriel
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Tracy Harrison
- Department of OB/GYN & Reproductive Sciences, University of California San Diego, San Diego, CA, United States
| | - Kirsten Kaptein
- Division of Plastic Surgery, University of California San Diego, San Diego, CA, United States
| | - Sanghee Lee
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Anne Z. Murphy
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Engy Said
- Division of Regional Anesthesia, University of California San Diego, San Diego, CA, United States
| | - Stacey Abigail Smith
- Division of Infection Disease, The Hope Clinic of Emory University, Atlanta, GA, United States
| | - David A. Thomas
- Office of Research on Women's Health, National Institutes of Health, Bethesda, MD, United States
| | - Maria del C. Valdés Hernández
- Department of Neuroimaging Sciences, Center for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor Trasvina
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Ursula Wesselmann
- Departments of Anesthesiology and Perioperative Medicine/Division of Pain Medicine, Neurology and Psychology, and Consortium for Neuroengineering and Brain-Computer Interfaces, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Tony L. Yaksh
- Department of Anesthesiology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
2
|
Muscogiuri G, Verde L, Vetrani C, Barrea L, Savastano S, Colao A. Obesity: a gender-view. J Endocrinol Invest 2024; 47:299-306. [PMID: 37740888 PMCID: PMC10859324 DOI: 10.1007/s40618-023-02196-z] [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: 03/16/2023] [Accepted: 09/08/2023] [Indexed: 09/25/2023]
Abstract
PURPOSE There is a growing awareness of the importance of understanding gender differences in obesity. The aim of this short review was to revise the current evidence on anthropometric characteristics and nutritional and pharmacological aspects of obesity from a gender perspective. METHODS A literature search within PubMed was performed. Selected publications related to obesity and gender differences were reviewed. RESULTS The prevalence of obesity among men is higher than in women, but women have a higher percentage of body fat content compared to men, and gender appears to be an important factor in the manifestation of central (android) or peripheral (gynoid) obesity. In addition, while in most clinical trials, women are still underrepresented, in clinical registration trials of anti-obesity drugs, women are commonly up-represented and gender-specific analysis is uncommon. Considering that adipose tissue is one of the factors affecting the volume of distribution of many drugs, mainly lipophilic drugs, gender differences might be expected in the pharmacokinetics and pharmacodynamics of anti-obesity drugs. Indeed, although Liraglutide 3 mg, a long-acting glucagon-like peptide-1 receptor agonist, and naltrexone/bupropion display lipophilic properties, currently, a gender-dose adjustment for both these drugs administration is not recommended. In addition, despite that predicted responders to treatment offer substantial opportunities for efficient use, especially of expensive new therapies, such as anti-obesity drugs, data on gender differences to identify early responders to both these have not yet been investigated. Finally, bariatric surgery gender disparity reflects healthcare practices. Weight loss similar, but differing effects: women need more correction and face psychology challenges; men have worse physiology and fewer comorbidity improvements. CONCLUSION Gender differences exist in obesity prevalence and phenotype, body fat distribution, drug efficacy, clinical trial representation, and different secondary effects of bariatric surgery. Gender is an important variable in obesity analysis.
Collapse
Affiliation(s)
- G Muscogiuri
- Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, Italy.
| | - L Verde
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - C Vetrani
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento Di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via PorzioIsola F2, 80143, Naples, Italy
| | - L Barrea
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento Di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via PorzioIsola F2, 80143, Naples, Italy
| | - S Savastano
- Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - A Colao
- Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Centro Italiano Per La Cura E Il Benessere del Paziente Con Obesità (C.I.B.O), Dipartimento Di Medicina Clinica E Chirurgia, Diabetologia E Andrologia, Unità Di Endocrinologia, Università Degli Studi Di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, Naples, Italy
| |
Collapse
|
3
|
Estrogen as a key regulator of energy homeostasis and metabolic health. Biomed Pharmacother 2022; 156:113808. [DOI: 10.1016/j.biopha.2022.113808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/02/2022] [Accepted: 10/03/2022] [Indexed: 11/23/2022] Open
|
4
|
Sayers S, Le N, Hernandez J, Mata‐Pacheco V, Wagner EJ. The vital role of arcuate nociceptin/orphanin FQ neurones in mounting an oestradiol-dependent adaptive response to negative energy balance via inhibition of nearby proopiomelanocortin neurones. J Physiol 2022; 600:4939-4961. [PMID: 36217719 PMCID: PMC9828807 DOI: 10.1113/jp283378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 09/28/2022] [Indexed: 01/12/2023] Open
Abstract
We tested the hypothesis that N/OFQ neurones in the arcuate nucleus (N/OFQARC ) inhibit proopiomelanocortin (POMCARC ) neurones in a diet- and hormone-dependent manner to promote a more extensive rebound hyperphagia upon re-feeding following an 18 h fast. We utilized intact male or ovariectomized (OVX) female mice subjected to ad libitum-feeding or fasting conditions. N/OFQARC neurones under negative energy balance conditions displayed heightened sensitivity as evidenced by a decreased rheobase threshold, increased firing frequency, and increased burst duration and frequency compared to ad libitum-feeding conditions. Stimulation of N/OFQARC neurones more robustly inhibited POMCARC neurones under fasting conditions compared to ad libitum-feeding conditions. N/OFQARC inhibition of POMCARC neurones is hormone dependent as chemostimulation of N/OFQARC neurones from fasted males and OVX females produced a sizable outward current in POMCARC neurones. Oestradiol (E2 ) markedly attenuated the N/OFQ-induced POMCARC outward current. Additionally, N/OFQ tonically inhibits POMCARC neurones to a greater degree under fasting conditions than in ad libitum-feeding conditions as evidenced by the abrogation of N/OFQ-nociceptin opioid peptide (NOP) receptor signalling and inhibition of N/OFQ release via chemoinhibition of N/OFQARC neurones. Intra-arcuate nucleus application of N/OFQ further elevated the hyperphagic response and increased meal size during the 6 h re-feed period, and these effects were mimicked by chemostimulation of N/OFQARC neurones in vivo. E2 attenuated the robust N/OFQ-induced rebound hyperphagia seen in vehicle-treated OVX females. These data demonstrate that N/OFQARC neurones play a vital role in mitigating the impact of negative energy balance by inhibiting the excitability of anorexigenic neural substrates, an effect that is diminished by E2 in females. KEY POINTS: Nociceptin/orphanin FQ (N/OFQ) promotes increased energy intake and decreased energy expenditure under conditions of positive energy balance in a sex- and hormone-dependent manner. Here it is shown that under conditions of negative energy balance, i.e. fasting, N/OFQ inhibits anorexigenic proopiomelanocortin (POMC) neurones to a greater degree compared to homeostatic conditions due to fasting-induced hyperexcitability of N/OFQ neurones. Additionally, N/OFQ promotes a sustained increase in rebound hyperphagia and increase in meal size during the re-feed period following a fast. These results promote greater understanding of how energy balance influences the anorexigenic circuitry of the hypothalamus, and aid in understanding the neurophysiological pathways implicated in eating disorders promoting cachexia.
Collapse
Affiliation(s)
- Sarah Sayers
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Nikki Le
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Jennifer Hernandez
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Veronica Mata‐Pacheco
- Graduate College of Biomedical SciencesWestern University of Health SciencesPomonaCAUSA
| | - Edward J. Wagner
- College of Osteopathic Medicine of the PacificWestern University of Health SciencesPomonaCAUSA
| |
Collapse
|
5
|
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: 2] [Impact Index Per Article: 1.0] [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
|
6
|
Balthazart J. Membrane-initiated actions of sex steroids and reproductive behavior: A historical account. Mol Cell Endocrinol 2021; 538:111463. [PMID: 34582978 DOI: 10.1016/j.mce.2021.111463] [Citation(s) in RCA: 7] [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: 05/04/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 01/25/2023]
Abstract
It was assumed for a long time that sex steroids are activating reproductive behaviors by the same mechanisms that produce their morphological and physiological effects in the periphery. However during the last few decades an increasing number of examples were identified where behavioral effects of steroids were just too fast to be mediated via changes in DNA transcription. This progressively forced behavioral neuroendocrinologists to recognize that part of the effects of steroids on behavior are mediated by membrane-initiated events. In this review we present a selection of these early data that changed the conceptual landscape and we provide a summary the different types of membrane-associated receptors (estrogens, androgens and progestagens receptors) that are playing the most important role in the control of reproductive behaviors. Then we finally describe in more detail three separate behavioral systems in which membrane-initiated events have clearly been established to contribute to behavior control.
Collapse
|
7
|
Ellis SN, Honeycutt JA. Sex Differences in Affective Dysfunction and Alterations in Parvalbumin in Rodent Models of Early Life Adversity. Front Behav Neurosci 2021; 15:741454. [PMID: 34803622 PMCID: PMC8600234 DOI: 10.3389/fnbeh.2021.741454] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
The early life environment markedly influences brain and behavioral development, with adverse experiences associated with increased risk of anxiety and depressive phenotypes, particularly in females. Indeed, early life adversity (ELA) in humans (i.e., caregiver deprivation, maltreatment) and rodents (i.e., maternal separation, resource scarcity) is associated with sex-specific emergence of anxious and depressive behaviors. Although these disorders show clear sex differences in humans, little attention has been paid toward evaluating sex as a biological variable in models of affective dysfunction; however, recent rodent work suggests sex-specific effects. Two widely used rodent models of ELA approximate caregiver deprivation (i.e., maternal separation) and resource scarcity (i.e., limited bedding). While these approaches model aspects of ELA experienced in humans, they span different portions of the pre-weaning developmental period and may therefore differentially contribute to underlying mechanistic risk. This is borne out in the literature, where evidence suggests differences in trajectories of behavior depending on the type of ELA and/or sex; however, the neural underpinning of these differences is not well understood. Because anxiety and depression are thought to involve dysregulation in the balance of excitatory and inhibitory signaling in ELA-vulnerable brain regions (e.g., prefrontal cortex, amygdala, hippocampus), outcomes are likely driven by alterations in local and/or circuit-specific inhibitory activity. The most abundant GABAergic subtypes in the brain, accounting for approximately 40% of inhibitory neurons, contain the calcium-binding protein Parvalbumin (PV). As PV-expressing neurons have perisomatic and proximal dendritic targets on pyramidal neurons, they are well-positioned to regulate excitatory/inhibitory balance. Recent evidence suggests that PV outcomes following ELA are sex, age, and region-specific and may be influenced by the type and timing of ELA. Here, we suggest the possibility of a combined role of PV and sex hormones driving differences in behavioral outcomes associated with affective dysfunction following ELA. This review evaluates the literature across models of ELA to characterize neural (PV) and behavioral (anxiety- and depressive-like) outcomes as a function of sex and age. Additionally, we detail a putative mechanistic role of PV on ELA-related outcomes and discuss evidence suggesting hormone influences on PV expression/function which may help to explain sex differences in ELA outcomes.
Collapse
Affiliation(s)
- Seneca N Ellis
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States
| | - Jennifer A Honeycutt
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States.,Department of Psychology, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
8
|
Ram A, Edwards TM, McCarty A, McDermott MV, Bobeck EN. Morphine-induced kinase activation and localization in the periaqueductal gray of male and female mice. J Neurochem 2021; 159:590-602. [PMID: 34499746 DOI: 10.1111/jnc.15506] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/19/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022]
Abstract
Morphine is a potent opioid analgesic with high propensity for the development of antinociceptive tolerance. Morphine antinociception and tolerance are partially regulated by the midbrain ventrolateral periaqueductal gray (vlPAG). However, the majority of research evaluating mu-opioid receptor signaling has focused on males. Here, we investigate kinase activation and localization patterns in the vlPAG following acute and chronic morphine treatment in both sexes. Male and female mice developed rapid antinociceptive tolerance to morphine (10 mg/kg i.p.) on the hot plate assay, but tolerance did not develop in males on the tail flick assay. Quantitative fluorescence immunohistochemistry was used to map and evaluate the activation of extracellular signal-regulated kinase 1/2 (ERK 1/2), protein kinase-C (PKC), and protein kinase-A (PKA). We observed significantly greater phosphorylated ERK 1/2 in the vlPAG of chronic morphine-treated animals which co-localized with the endosomal marker, Eea1. We note that pPKC is significantly elevated in the vlPAG of both sexes following chronic morphine treatment. We also observed that although PKA activity is elevated following chronic morphine treatment in both sexes, there is a significant reduction in the nuclear translocation of its phosphorylated substrate. Taken together, this study demonstrates increased activation of ERK 1/2, PKC, and PKA in response to repeated morphine treatment. The study opens avenues to explore the impact of chronic morphine treatment on G-protein signaling and kinase nuclear transport.
Collapse
Affiliation(s)
- Akila Ram
- Department of Biology, Utah State University, Logan, Utah, USA
| | | | - Ashley McCarty
- Department of Biology, Utah State University, Logan, Utah, USA
| | - Max V McDermott
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| | - Erin N Bobeck
- Department of Biology, Utah State University, Logan, Utah, USA
- Interdisciplinary Neuroscience Program, Utah State University, Logan, Utah, USA
| |
Collapse
|
9
|
G-Protein-Coupled Estrogen Receptor (GPER) in the Rostral Ventromedial Medulla Is Essential for Mobilizing Descending Inhibition of Itch. J Neurosci 2021; 41:7727-7741. [PMID: 34349001 DOI: 10.1523/jneurosci.2592-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 07/19/2021] [Accepted: 07/28/2021] [Indexed: 11/21/2022] Open
Abstract
Chronic itch is a troublesome condition and often difficult to cure. Emerging evidence suggests that the periaqueductal gray (PAG)-rostral ventromedial medulla (RVM) pathway may play an important role in the regulation of itch, but the cellular organization and molecular mechanisms remain incompletely understood. Here, we report that a group of RVM neurons distinctively express the G-protein-coupled estrogen receptor (GPER), which mediates descending inhibition of itch. We found that GPER+ neurons in the RVM were activated in chronic itch conditions in rats and mice. Selective ablation or chemogenetic suppression of RVM GPER+ neurons resulted in mechanical alloknesis and increased scratching in response to pruritogens, whereas chemogenetic activation of GPER+ neurons abrogated itch responses, indicating that GPER+ neurons are antipruritic. Moreover, GPER-deficient mice and rats of either sex exhibited hypersensitivity to mechanical and chemical itch, a phenotype reversible by the µ type opioid receptor (MOR) antagonism. Additionally, significant MOR phosphorylation in the RVM was detected in chronic itch models in wild-type but not in GPER-/- rats. Therefore, GPER not only identifies a population of medullary antipruritic neurons but may also determine the descending antipruritic tone through regulating µ opioid signaling.SIGNIFICANCE STATEMENT Therapeutic options for itch are limited because of an as yet incomplete understanding of the mechanisms of itch processing. Our data have provided novel insights into the cellular organization and molecular mechanisms of descending regulation of itch in normal and pathologic conditions. GPER+ neurons (largely GABAergic) in the RVM are antipruritic neurons under tonic opioidergic inhibition, activation of GPER promotes phosphorylation of MOR and disinhibition of the antipruritic GPER+ neurons from inhibitory opioidergic inputs, and failure to mobilize GPER+ neurons may result in the exacerbation of itch. Our data also illuminate on some of the outstanding questions in the field, such as the mechanisms underlying sex bias in itch, pain, and opioid analgesia and the paradoxical effects of morphine on pain and itch.
Collapse
|
10
|
Chae SA, Son JS, Du M. Prenatal exercise in fetal development: a placental perspective. FEBS J 2021; 289:3058-3071. [PMID: 34449982 DOI: 10.1111/febs.16173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/09/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Maternal obesity (MO) and gestational diabetes mellitus (GDM) are common in Western societies, which impair fetal development and predispose offspring to metabolic dysfunction. Placenta is the organ linking the mother to her fetus, and MO suppresses the development of vascular system and expression of nutrient transporters in placenta, thereby affecting fetal development. For maintaining its proper physiological function, placenta is energy demanding, which is met through extensive oxidative phosphorylation. However, the oxidative capacity of placenta is suppressed due to MO and GDM. Recently, several studies showed that physical activity during pregnancy enhances oxidative metabolism and improves placental function, which might be partially mediated by exerkines, referring to cytokines elicited by exercise. In addition, as an endocrine organ, placenta secretes cytokines, termed placentokines, including apelin, superoxide dismutase 3, irisin, and adiponectin, which mediate fetal development and maternal metabolism. Possible molecular mechanisms linking maternal exercise and placentokines to placental and fetal development are further discussed. As an emerging field, up to now, available studies are limited, mostly conducted in rodents. Given the epidemics of obesity and metabolic disorders, as well as the prevalence of maternal sedentary lifestyle, the effects of exercise of pregnant women on placental function and placentokine secretion, as well as their impacts on fetal development, need to be further examined.
Collapse
Affiliation(s)
- Song Ah Chae
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Jun Seok Son
- Laboratory of Perinatal Kinesioepigenetics, Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Min Du
- Nutrigenomics and Growth Biology Laboratory, Department of Animal Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
11
|
McHann MC, Blanton HL, Guindon J. Role of sex hormones in modulating breast and ovarian cancer associated pain. Mol Cell Endocrinol 2021; 533:111320. [PMID: 34033890 PMCID: PMC8263503 DOI: 10.1016/j.mce.2021.111320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/16/2021] [Accepted: 05/09/2021] [Indexed: 01/18/2023]
Abstract
According to the National Cancer Institute in 2020 there will be an estimated 21,750 new ovarian cancer cases and 276,480 new breast cancer cases. Both breast and ovarian cancer are hormone dependent cancers, meaning they cannot grow without the presence of hormones. The two most studied hormones in these two cancers are estrogen and progesterone, which are also involved in the modulation of pain. The incidence of pain in breast and ovarian cancer is very high. Research about mechanisms involved in modulation of pain by hormones are still being debated, as some studies find estrogen to be anti-nociceptive and others pro-nociceptive in pain studies. Moreover, analgesic treatments for breast and ovarian cancer-associated pain are limited and often ineffective. In this review, we will focus on estrogen and progesterone mechanisms of action in modulation of pain and cancer. We will also discuss new treatment options for these types of cancer and associated-pain.
Collapse
Affiliation(s)
- Melissa C McHann
- Department of Pharmacology and Neuroscience at Texas Tech University Health Sciences Center, USA
| | - Henry L Blanton
- Department of Pharmacology and Neuroscience at Texas Tech University Health Sciences Center, USA
| | - Josée Guindon
- Department of Pharmacology and Neuroscience at Texas Tech University Health Sciences Center, USA.
| |
Collapse
|
12
|
Xu JJ, Gao P, Wu Y, Yin SQ, Zhu L, Xu SH, Tang D, Cheung CW, Jiao YF, Yu WF, Li YH, Yang LQ. G protein-coupled estrogen receptor in the rostral ventromedial medulla contributes to the chronification of postoperative pain. CNS Neurosci Ther 2021; 27:1313-1326. [PMID: 34255932 PMCID: PMC8504531 DOI: 10.1111/cns.13704] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/19/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022] Open
Abstract
Aims Chronification of postoperative pain is a common clinical phenomenon following surgical operation, and it perplexes a great number of patients. Estrogen and its membrane receptor (G protein‐coupled estrogen receptor, GPER) play a crucial role in pain regulation. Here, we explored the role of GPER in the rostral ventromedial medulla (RVM) during chronic postoperative pain and search for the possible mechanism. Methods and Results Postoperative pain was induced in mice or rats via a plantar incision surgery. Behavioral tests were conducted to detect both thermal and mechanical pain, showing a small part (16.2%) of mice developed into pain persisting state with consistent low pain threshold on 14 days after incision surgery compared with the pain recovery mice. Immunofluorescent staining assay revealed that the GPER‐positive neurons in the RVM were significantly activated in pain persisting rats. In addition, RT‐PCR and immunoblot analyses showed that the levels of GPER and phosphorylated μ‐type opioid receptor (p‐MOR) in the RVM of pain persisting mice were apparently increased on 14 days after incision surgery. Furthermore, chemogenetic activation of GPER‐positive neurons in the RVM of Gper‐Cre mice could reverse the pain threshold of pain recovery mice. Conversely, chemogenetic inhibition of GPER‐positive neurons in the RVM could prevent mice from being in the pain persistent state. Conclusion Our findings demonstrated that the GPER in the RVM was responsible for the chronification of postoperative pain and the downstream pathway might be involved in MOR phosphorylation.
Collapse
Affiliation(s)
- Jia-Jia Xu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Po Gao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ying Wu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Su-Qing Yin
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Sai-Hong Xu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Chi-Wai Cheung
- Department of Anesthesiology, The University of Hong Kong, Hong Kong, China
| | - Ying-Fu Jiao
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yuan-Hai Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li-Qun Yang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
13
|
Lopresti NM, Esguerra M, Mermelstein PG. Sex Differences in Animal Models of Opioid Reward. CURRENT SEXUAL HEALTH REPORTS 2020; 12:186-194. [PMID: 33574737 PMCID: PMC7872138 DOI: 10.1007/s11930-020-00266-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss sex differences observed in preclinical rodent models of opioid reward. RECENT FINDINGS Utilizing a variety of methodological approaches and drug regimens, no clear consensus has emerged regarding the effects of opiates between males and females. This is quite dissimilar to work examining psychostimulants, where female animals reliably exhibit stronger behavioral responses. SUMMARY With opioid research quickly expanding to determine the neural underpinnings of opioid addiction, further research is essential to determine the conditions wherein sex differences may occur and how they may relate to the human condition.
Collapse
Affiliation(s)
| | - Manuel Esguerra
- University of Minnesota, Twin Cities, Department of Neuroscience
| | | |
Collapse
|
14
|
Ding X, Gao T, Gao P, Meng Y, Zheng Y, Dong L, Luo P, Zhang G, Shi X, Rong W. Activation of the G Protein-Coupled Estrogen Receptor Elicits Store Calcium Release and Phosphorylation of the Mu-Opioid Receptors in the Human Neuroblastoma SH-SY5Y Cells. Front Neurosci 2019; 13:1351. [PMID: 31920512 PMCID: PMC6928052 DOI: 10.3389/fnins.2019.01351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022] Open
Abstract
Estrogens exert extensive influences on the nervous system besides their well-known roles in regulation of reproduction and metabolism. Estrogens act via the nuclear receptor ERα and ERβ to regulate gene transcription (classical genomic effects). In addition, estrogens are also known to cause rapid non-genomic effects on neuronal functions including inducing fast changes in cytosolic calcium level and rapidly desensitizing the μ type opioid receptor (MOR). The receptors responsible for the rapid actions of estrogens remain uncertain, but recent evidence points to the G protein-coupled estrogen receptor (GPER), which has been shown to be expressed widely in the nervous system. In the current study, we test the hypothesis that activation of GPER may mediate rapid calcium signaling, which may promote phosphorylation of MOR through the calcium-dependent protein kinases in neuronal cells. By qPCR and immunocytochemistry, we found that the human neuroblastoma SH-SY5Y cells endogenously express GPER and MOR. Activation of GPER by 17β-estradiol (E2) and G-1 (GPER selective agonist) evoked a rapid calcium rise in a concentration-dependent manner, which was due to store release rather than calcium entry. The GPER antagonist G15, the PLC inhibitor U73122 and the IP3 receptor inhibitor 2-APB each virtually abolished the calcium responses to E2 or G-1. Activation of GPER stimulated translocation of PKC isoforms (α and ε) to the plasma membrane, which led to MOR phosphorylation. Additionally, E2 and G-1 stimulated c-Fos expression in SH-SY5Y cells in a PLC/IP3-dependent manner. In conclusion, the present study has revealed a novel GPER-mediated estrogenic signaling in neuroblastoma cells in which activation of GPER is followed by rapid calcium mobilization, PKC activation and MOR phosphorylation. GPER-mediated rapid calcium signal may also be transmitted to the nucleus to impact on gene transcription. Such signaling cascade may play important roles in the regulation of opioid signaling in the brain.
Collapse
Affiliation(s)
- Xiaowei Ding
- Department of Anesthesiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Gao
- Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Po Gao
- Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Youqiang Meng
- Department of Neurosurgery, Xin Hua Hospital Chongming Branch, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Zheng
- Department of Anesthesiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Dong
- Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Luo
- Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guohua Zhang
- Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyin Shi
- Department of Anesthesiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weifang Rong
- Department of Anesthesiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Anatomy and Physiology, Faculty of Basic Medical Sciences, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Domínguez-Ordóñez R, García-Juárez M, Lima-Hernández FJ, Gómora-Arrati P, Domínguez-Salazar E, Luna-Hernández A, Hoffman KL, Blaustein JD, Etgen AM, González-Flores O. Protein kinase inhibitors infused intraventricularly or into the ventromedial hypothalamus block short latency facilitation of lordosis by oestradiol. J Neuroendocrinol 2019; 31:e12809. [PMID: 31715031 DOI: 10.1111/jne.12809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 11/27/2022]
Abstract
An injection of unesterified oestradiol (E2 ) facilitates receptive behaviour in E2 benzoate (EB)-primed, ovariectomised female rats when it is administered i.c.v. or systemically. The present study tested the hypothesis that inhibitors of protein kinase A (PKA), protein kinase G (PKG) or the Src/mitogen-activated protein kinase (MAPK) complex interfere with E2 facilitation of receptive behaviour. In Experiment 1, lordosis induced by i.c.v. infusion of E2 was significantly reduced by i.c.v. administration of Rp-cAMPS, a PKA inhibitor, KT5823, a PKG inhibitor, and PP2 and PD98059, Src and MAPK inhibitors, respectively, between 30 and 240 minutes after infusion. In Experiment 2, we determined whether the ventromedial hypothalamus (VMH) is one of the neural sites at which those intracellular pathways participate in lordosis behaviour induced by E2 . Administration of each of the four protein kinase inhibitors into the VMH blocked facilitation of lordosis induced by infusion of E2 also into the VMH. These data support the hypothesis that activation of several protein kinase pathways is involved in the facilitation of lordosis by E2 in EB-primed rats.
Collapse
Affiliation(s)
- Raymundo Domínguez-Ordóñez
- Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Tecamachalco, Mexico
- Department of Psychological and Brain Sciences, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA, USA
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Francisco J Lima-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Porfirio Gómora-Arrati
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Emilio Domínguez-Salazar
- Área de Neurociencias, Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, México
| | - Ailyn Luna-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Kurt L Hoffman
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
| | - Jeffrey D Blaustein
- Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Tecamachalco, Mexico
- Department of Psychological and Brain Sciences, Center for Neuroendocrine Studies, University of Massachusetts, Amherst, MA, USA
| | - Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, México
- Área de Neurociencias, Departamento de Biología de la Reproducción, Universidad Autónoma Metropolitana, Iztapalapa, México
| |
Collapse
|
16
|
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: 2.8] [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
|
17
|
Cataldi M, Muscogiuri G, Savastano S, Barrea L, Guida B, Taglialatela M, Colao A. Gender-related issues in the pharmacology of new anti-obesity drugs. Obes Rev 2019; 20:375-384. [PMID: 30589980 DOI: 10.1111/obr.12805] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 10/01/2018] [Accepted: 10/13/2018] [Indexed: 12/14/2022]
Abstract
Four new medicines-liraglutide, lorcaserin, bupropion/naltrexone, and phentermine/topiramate-have been recently added to the pharmacological arsenal for obesity treatment and could represent important tools to manage this epidemic disease. To achieve satisfactory anti-obesity goals, the use of these new medicines should be optimized and tailored to specific patient subpopulations also by applying dose adjustments if needed. In the present review, we posit that gender could be among the factors influencing the activity of the new obesity drugs both because of pharmacokinetic and pharmacodynamic factors. Although evidence from premarketing clinical studies suggested that no dose adjustment by gender is necessary for any of these new medicines, these studies were not specifically designed to identify gender-related differences. This observation, together with the strong theoretical background supporting the hypothesis of a gender-dimorphic response, strongly call upon an urgent need of new real-life data on gender-related difference in the pharmacology of these new obesity drugs.
Collapse
Affiliation(s)
- Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Giovanna Muscogiuri
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Silvia Savastano
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Luigi Barrea
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Bruna Guida
- Division of Physiology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| | - Annamaria Colao
- Division of Endocrinology, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy.,Federico II University Hospital, Naples, Italy
| |
Collapse
|
18
|
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: 4.8] [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
|
19
|
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: 5.7] [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
|
20
|
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: 19] [Impact Index Per Article: 3.2] [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
|
21
|
Inhibitor of Differentiation-3 and Estrogenic Endocrine Disruptors: Implications for Susceptibility to Obesity and Metabolic Disorders. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6821601. [PMID: 29507860 PMCID: PMC5817379 DOI: 10.1155/2018/6821601] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 11/07/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022]
Abstract
The rising global incidence of obesity cannot be fully explained within the context of traditional risk factors such as an unhealthy diet, physical inactivity, aging, or genetics. Adipose tissue is an endocrine as well as a metabolic organ that may be susceptible to disruption by environmental estrogenic chemicals. Since some of the endocrine disruptors are lipophilic chemicals with long half-lives, they tend to bioaccumulate in the adipose tissue of exposed populations. Elevated exposure to these chemicals may predispose susceptible individuals to weight gain by increasing the number and size of fat cells. Genetic studies have demonstrated that the transcriptional regulator inhibitor of differentiation-3 (ID3) promotes high fat diet-induced obesity in vivo. We have shown previously that PCB153 and natural estrogen 17β-estradiol increase ID3 expression. Based on our findings, we postulate that ID3 is a molecular target of estrogenic endocrine disruptors (EEDs) in the adipose tissue and a better understanding of this relationship may help to explain how EEDs can lead to the transcriptional programming of deviant fat cells. This review will discuss the current understanding of ID3 in excess fat accumulation and the potential for EEDs to influence susceptibility to obesity or metabolic disorders via ID3 signaling.
Collapse
|
22
|
Gao P, Ding XW, Dong L, Luo P, Zhang GH, Rong WF. Expression of aromatase in the rostral ventromedial medulla and its role in the regulation of visceral pain. CNS Neurosci Ther 2017; 23:980-989. [PMID: 29047208 DOI: 10.1111/cns.12769] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 12/30/2022] Open
Abstract
AIMS Estrogens are known to exert a wide spectrum of actions on brain functions including modulation of pain. Besides the circulating estrogens produced mainly by the ovaries, many brain regions are also capable of de novo synthesizing estrogens, which may exert important modulatory effects on neuronal functions. This study was aimed to test the hypothesis that aromatase, the enzyme that catalyzes the conversion of testosterone to estradiols, may be distributed in the rostral ventromedial medulla (RVM), where it may impact on visceral pain. METHODS AND RESULTS Adult female rats were treated with cyclophosphamide (CPM, 50 mg/kg, ip, once every 3 days) or saline. At approximately day 10 following the 3rd injection, CPM-treated rats exhibited colorectal hyperalgesia as they showed significantly greater abdominal withdrawal responses (AWR) to graded colorectal distension (CRD, 0-100 mm Hg) than the saline group. Immunofluorescent staining and Western blot assay revealed that CPM-induced colorectal hyperalgesia was associated with significantly increased expression of aromatase and phosphorylated μ-type opioid receptor (pMOR) and decreased expression of total MOR in the RVM. Intracisternal application of aromatase inhibitors, fadrozole, and letrozole reversed CPM-induced colorectal hyperalgesia and restored pMOR and MOR expression in the RVM. CONCLUSIONS Our observations confirmed the expression of aromatase in the RVM, a pivotal brain region in descending modulation of pain and opioid analgesia. The results support the hypothesis that locally produced estrogens in the RVM may be involved in the maintenance of chronic visceral hyperalgesia and the downstream signaling may involve phosphorylation of MOR.
Collapse
Affiliation(s)
- Po Gao
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao-Wei Ding
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Li Dong
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ping Luo
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guo-Hua Zhang
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Fang Rong
- Hongqiao International Institute of Medical Research, Tongren Hospital and Department of Physiology, Faculty of Basic Medical Sciences, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
23
|
Chiang YC, Wang RY, Huang CL, Chen SH, Ho WJ, Lane HY, Ho IK, Yang HT, Ma WL. Reduced dosing and liability in methadone maintenance treatment by targeting oestrogen signal for morphine addiction. J Cell Mol Med 2017; 21:3552-3564. [PMID: 28699698 PMCID: PMC5706516 DOI: 10.1111/jcmm.13266] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 05/02/2017] [Indexed: 11/29/2022] Open
Abstract
Methadone maintenance treatment (MMT) is the major tapering therapy for morphine addictive patients. There have gender differences reported in response to MMT. This study discovered that the estrogen‐response element single nucleotide polymorphism (ERE‐SNP; rs16974799, C/T) of cytochrome 2B6 gene (cyp2b6; methadone catabolic enzyme) responded differently to MMT dosing. Oestradiol was associated with high MMT dosing, high enantiomer (R‐ or S‐) of 2‐ethylidene‐1,5‐dimethyl‐3,3‐dipheny‐pyrrolidine (EDDP; methadone metabolite) to methadone ratio and increased drug‐seeking behaviour, implicating oestradiol‐CYP‐EDDP/methadone axis decreasing MMT efficacy. In mouse model, oestrogen mitigates methadone antinociceptive response, facilitates methadone catabolism and up‐regulates methadone‐associated metabolizing enzymes. Oestrogen also ablates chronic methadone administration‐induced rewarding response. Mechanism dissection revealed the CC genotype of CYP2B6‐ERE‐SNP exerts higher ERE sequence alignment score, higher estrogenic response as compared to TT genotype. At last, preclinical study via targeting estrogen signal that tamoxifen (TMX; selective estrogen receptor modulator, SERM) could facilitate the tolerance phase rewarding response of methadone. Strikingly, TMX also reduces tapering/abstinence phases methadone liability in mice. In conclusion, this study demonstrates altering methadone metabolism through targeting estrogen signals might be able to free morphine addictive patients from the addiction of opioid replacement therapy. Therefore, the add‐on therapy clinical trial introducing SERM in MMT regimen is suggested.
Collapse
Affiliation(s)
- Yao-Chang Chiang
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan.,Department of Nursing, Division of Basic Medical Sciences, Chang Gong University of Science and Technology, Chiayi County, Taiwan
| | - Ruey-Yun Wang
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan.,Department of Public Health, China Medical University, Taichung, Taiwan
| | - Chieh-Liang Huang
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan.,Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Shue-Hwa Chen
- Sex Hormone Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Jing Ho
- Sex Hormone Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Hsien-Yuan Lane
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan.,Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan.,Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Ing-Kang Ho
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan
| | - Hwei-Ting Yang
- Graduate Institution of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Wen-Lung Ma
- Center for Drug Abuse and Addiction, Brain Disease Research Center, China Medical University, Taichung, Taiwan.,Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan.,Sex Hormone Research Center, China Medical University Hospital, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
24
|
Gómora-Arrati P, Dominguez G, Ågmo A. GABA Receptors in the Medial Preoptic Area Modulate the Onset of Oestradiol-Induced Maternal Behaviour in Hysterectomised-Ovariectomised, Pregnant Rats. J Neuroendocrinol 2016; 28. [PMID: 27631525 DOI: 10.1111/jne.12434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 09/01/2016] [Accepted: 09/12/2016] [Indexed: 11/30/2022]
Abstract
We studied the participation of GABA neurotransmission in the medial preoptic area (mPOA) with respect to the onset of the pup retrieval response and nest building. Pregnant female rats were implanted with bilateral cannulae in the mPOA on day 12 of pregnancy and, on day 16, the females were hysterectomised and ovariectomised and given 200 μg/kg of oestradiol benzoate. Two days later, the females received one of the following intracerebral drug treatments: GABAB agonist baclofen (200 ng); GABAB antagonist phaclofen (1 μg); GABAA antagonist bicuculline (60 ng); or physiological saline. Five minutes after intracerebral infusion, three foster pups were introduced into the females' home cage. The subjects were observed for pup grouping (retrieval) during 15 min, after which the pups were left with the female. During the next 12 h, an observation was made every 1 h to determine whether the pups had been grouped (retrieved) or not. The GABAB agonist baclofen reduced the proportion of females retrieving pups from 4 to 8 h following pup introduction. By contrast, both the GABAA antagonist bicuculline and the GABAB antagonist phaclofen enhanced the proportion of females retrieving pups during the first 3 h of observation. The latency to pup retrieval in subjects treated with the GABAB agonist baclofen was significantly longer than that in subjects given any of the antagonists. All females built a nest but baclofen reduced nest quality. These data show that activation of GABAB receptors in the mPOA has an inhibitory effect on basic maternal behaviours, whereas blockade of either the GABAA or GABAB receptor facilitates pup retrieval. It is possible that reduced GABAergic tone in the mPOA is a key element in the initiation of maternal behaviours in postparturient rats.
Collapse
Affiliation(s)
- P Gómora-Arrati
- Centro de Investigación en Reproducción Animal, CINVESTAV-UAT, Tlaxcala, Mexico
| | - G Dominguez
- Centro de Investigación en Reproducción Animal, CINVESTAV-UAT, Tlaxcala, Mexico
| | - A Ågmo
- Department of Psychology, University of Tromsø, Tromsø, Norway
| |
Collapse
|
25
|
Domínguez-Ordóñez R, García-Juárez M, Lima-Hernández FJ, Gómora-Arrati P, Blaustein JD, Etgen AM, González-Flores O. Estrogen receptor α and β are involved in the activation of lordosis behavior in estradiol-primed rats. Horm Behav 2016; 86:1-7. [PMID: 27594441 DOI: 10.1016/j.yhbeh.2016.08.010] [Citation(s) in RCA: 16] [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: 05/15/2016] [Revised: 08/28/2016] [Accepted: 08/31/2016] [Indexed: 11/17/2022]
Abstract
The present study was designed to assess the participation of estrogen receptors alpha (ERα) and beta (ERβ) in the short-term facilitation of lordosis behavior in ovariectomized (ovx), estradiol (E2) primed rats. In experiment 1, dose response curves for PPT and DPN (ERα and ERβ agonists, respectively) facilitation of lordosis behavior (lordosis quotient and lordosis score) were established by infusing these agonists into the right lateral ventricle (icv) in female rats injected 40h previously with 5μg of E2 benzoate. PPT doses of 0.08 and 0.4ng produced high lordosis quotients starting at 30min and continuing at 120 and 240min post-injection. DPN induced high levels of lordosis behavior at all times tested. However, the intensity of lordosis induced by both agonists was weak. In experiment 2, we tested the involvement of each ER in facilitation of lordosis by icv infusion of MPP (ERα-selective antagonist) or PHTPP (ERβ-selective antagonist) prior to infusion of 2ng of free E2. Icv infusion of either MPP or PHTPP 30min before free E2 significantly depressed E2 facilitation of lordosis. The results suggest that both forms of ER are involved in the short-latency facilitation of lordosis behavior in E2-primed rats.
Collapse
Affiliation(s)
- Raymundo Domínguez-Ordóñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México; Doctorado en Ciencias Biológicas, Universidad Autónoma de Tlaxcala, Tlaxcala, México
| | - Marcos García-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México
| | - Francisco J Lima-Hernández
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México; Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tlaxcala, México
| | - Porfirio Gómora-Arrati
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México
| | - Jeffrey D Blaustein
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Anne M Etgen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, México.
| |
Collapse
|
26
|
Acaz-Fonseca E, Avila-Rodriguez M, Garcia-Segura LM, Barreto GE. Regulation of astroglia by gonadal steroid hormones under physiological and pathological conditions. Prog Neurobiol 2016; 144:5-26. [DOI: 10.1016/j.pneurobio.2016.06.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 06/05/2016] [Indexed: 01/07/2023]
|
27
|
Kow LM, Pfaff DW. Rapid estrogen actions on ion channels: A survey in search for mechanisms. Steroids 2016; 111:46-53. [PMID: 26939826 PMCID: PMC4929851 DOI: 10.1016/j.steroids.2016.02.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 12/31/2022]
Abstract
A survey of nearly two hundred reports shows that rapid estrogenic actions can be detected across a range of kinds of estrogens, a range of doses, on a wide range of tissue, cell and ion channel types. Striking is the fact that preparations of estrogenic agents that do not permeate the cell membrane almost always mimic the actions of the estrogenic agents that do permeate the membrane. All kinds of estrogens, ranging from natural ones, through receptor modulators, endocrine disruptors, phytoestrogens, agonists, and antagonists to novel G-1 and STX, have been reported to be effective. For actions on specific types of ion channels, the possibility of opposing actions, in different cases, is the rule, not the exception. With this variety there is no single, specific action mechanism for estrogens per se, although in some cases estrogens can act directly or via some signaling pathways to affect ion channels. We infer that estrogens can bind a large number of substrates/receptors at the membrane surface. As against the variety of subsequent routes of action, this initial step of the estrogen's binding action is the key.
Collapse
Affiliation(s)
- Lee-Ming Kow
- The Rockefeller University, New York, NY 10065, USA.
| | | |
Collapse
|
28
|
Caldwell JD, Gebhart VM, Jirikowski GF. Estradiol's interesting life at the cell's plasma membrane. Steroids 2016; 111:4-11. [PMID: 27018128 DOI: 10.1016/j.steroids.2016.03.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/14/2016] [Indexed: 10/22/2022]
Abstract
Clearly, we have presented here evidence of a very complex set of mechanisms and proteins involved with various and intricate actions of steroids at the plasma membrane. Steroids do MUCH more at the plasma membrane than simply passing passively through it. They may sit in the membrane; they are bound by numerous proteins in the membrane, including ERs, SHBG, steroid-binding globulin receptors, and perhaps elements of cellular architecture such as tubulin. It also seems likely that the membrane itself responds graphically to the presence of steroids by actually changing its shape as well, perhaps, as accumulating steroids. Clara Szego suggested in the 1980s that actions of E2 at one level would act synergistically with its actions at another level (e.g. membrane actions would complement nuclear actions). Given the sheer number of proteins involved in steroid actions, just at the membrane level, it seems unlikely that every action of a steroid on every potential protein effector will act to the same end. It seems more likely that these multiple effects and sites of effect of steroids contribute to the confusion that exists as to what actions steroids always have. For example, there is confusion with regard to synthetic agents (SERMs etc.) that have different and often opposite actions depending on which organ they act upon. A better understanding of the basic actions of steroids should aid in understanding the variability of their clinical effects.
Collapse
Affiliation(s)
- J D Caldwell
- Edward Via College of Osteopathic Medicine, Dept. of Pharmacology, Spartanburg, SC, USA.
| | - V M Gebhart
- Jena University Hospital, Inst. Anatomie II, Jena, Germany
| | - G F Jirikowski
- Jena University Hospital, Inst. Anatomie II, Jena, Germany
| |
Collapse
|
29
|
Santollo J, Daniels D. Multiple estrogen receptor subtypes influence ingestive behavior in female rodents. Physiol Behav 2015; 152:431-7. [PMID: 26037634 DOI: 10.1016/j.physbeh.2015.05.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/27/2015] [Accepted: 05/28/2015] [Indexed: 11/24/2022]
Abstract
Postmenopausal women are at an increased risk of obesity and cardiovascular-related diseases. This is attributable, at least in part, to loss of the ovarian hormone estradiol, which inhibits food and fluid intake in humans and laboratory animal models. Although the hypophagic and anti-dipsogenic effects of estradiol have been well documented for decades, the precise mechanisms underlying these effects are not fully understood. An obvious step toward addressing this open question is identifying which estrogen receptor subtypes are involved and what intracellular processes are involved. This question, however, is complicated not only by the variety of estrogen receptor subtypes that exist, but also because many subtypes have multiple locations of action (i.e. in the nucleus or in the plasma membrane). This review will highlight our current understanding of the roles that specific estrogen receptor subtypes play in mediating estradiol's anorexigenic and anti-dipsogenic effects along with highlighting the many open questions that remain. This review will also describe recent work being performed by our laboratory aimed at answering these open questions.
Collapse
Affiliation(s)
- Jessica Santollo
- Department of Psychology, University at Buffalo, Buffalo, NY 14260, United States
| | - Derek Daniels
- Department of Psychology, University at Buffalo, Buffalo, NY 14260, United States.
| |
Collapse
|
30
|
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: 35] [Impact Index Per Article: 3.9] [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
|
31
|
Sinchak K, Dalhousay L, Sanathara N. Orphanin FQ-ORL-1 regulation of reproduction and reproductive behavior in the female. VITAMINS AND HORMONES 2015; 97:187-221. [PMID: 25677773 DOI: 10.1016/bs.vh.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Orphanin FQ (OFQ/N) and its receptor, opioid receptor-like receptor-1 (ORL-1), are expressed throughout steroid-responsive limbic and hypothalamic circuits that regulate female ovarian hormone feedback and reproductive behavior circuits. The arcuate nucleus of the hypothalamus (ARH) is a brain region that expresses OFQ/N and ORL-1 important for both sexual behavior and modulating estradiol feedback loops. Within the ARH, the activation of the OFQ/N-ORL-1 system facilitates sexual receptivity (lordosis) through the inhibition of β-endorphin neuronal activity. Estradiol initially activates ARH β-endorphin neurons to inhibit lordosis. Simultaneously, estradiol upregulates coexpression of OFQ/N and progesterone receptors and ORL-1 in ARH β-endorphin neurons. Ovarian hormones regulate pre- and postsynaptic coupling of ORL-1 to its G protein-coupled signaling pathways. When the steroid-primed rat is nonreceptive, estradiol acts pre- and postsynaptically to decrease the ability of the OFQ/N-ORL-1 system to inhibit ARH β-endorphin neurotransmission. Conversely, when sexually receptive, ORL-1 signaling is restored to inhibit β-endorphin neurotransmission. Although steroid signaling that facilitates lordosis converges to deactivate ARH β-endorphin neurons, estradiol-only facilitation of lordosis requires the activation of ORL-1, but estradiol+progesterone does not, indicating that multiple circuits mediate ovarian hormone signaling to deactivate ARH β-endorphin neurons. Research on the role of OFQ/N-ORL-1 in ovarian hormone feedback loops is just beginning. In the rat, OFQ/N may act to terminate gonadotropin-releasing hormone and luteinizing hormone release under positive and negative feedbacks. In the ewe, it appears to directly inhibit gonadotropin-releasing hormone release to mediate progesterone-negative feedback. As a whole, the localization and actions of OFQ/N-ORL-1 system indicate that it may mediate the actions of estradiol and progesterone to synchronize reproductive behavior and ovarian hormone feedback loops.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, California, USA.
| | - Lauren Dalhousay
- Department of Biological Sciences, California State University, Long Beach, California, USA
| | - Nayna Sanathara
- Department of Pharmacological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
32
|
Rønnekleiv OK, Zhang C, Bosch MA, Kelly MJ. Kisspeptin and Gonadotropin-Releasing Hormone Neuronal Excitability: Molecular Mechanisms Driven by 17β-Estradiol. Neuroendocrinology 2014; 102:184-93. [PMID: 25612870 PMCID: PMC4459938 DOI: 10.1159/000370311] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/02/2014] [Indexed: 11/19/2022]
Abstract
Kisspeptin is a neuropeptide that signals via a Gαq-coupled receptor, GPR54, in gonadotropin-releasing hormone (GnRH) neurons and is essential for pubertal maturation and fertility. Kisspeptin depolarizes and excites GnRH neurons primarily through the activation of canonical transient receptor potential (TRPC) channels and the inhibition of K+ channels. The gonadal steroid 17β-estradiol (E2) upregulates not only kisspeptin (Kiss1) mRNA but also increases the excitability of the rostral forebrain Kiss1 neurons. In addition, a primary postsynaptic action of E2 on GnRH neurons is to upregulate the expression of channel transcripts that orchestrate the downstream signaling of kisspeptin in GnRH neurons. These include not only TRPC4 channels but also low-voltage-activated T-type calcium channels and high-voltage-activated L-, N- and R-type calcium channel transcripts. Moreover, E2 has direct membrane-initiated actions to alter the excitability of GnRH neurons by enhancing ATP-sensitive potassium channel activity, which is critical for maintaining GnRH neurons in a hyperpolarized state for the recruitment of T-type calcium channels that are important for burst firing. Therefore, E2 modulates the excitability of GnRH neurons as well as of Kiss1 neurons by altering the expression and/or function of ion channels; moreover, kisspeptin provides critical excitatory input to GnRH neurons to facilitate burst firing activity and peptide release.
Collapse
Affiliation(s)
- Oline K. Rønnekleiv
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Chunguang Zhang
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Martha A. Bosch
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
| | - Martin J. Kelly
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, Oregon
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, USA
| |
Collapse
|
33
|
Teodorov E, Camarini R, Bernardi M, Felicio L. Treatment with steroid hormones and morphine alters general activity, sexual behavior, and opioid gene expression in female rats. Life Sci 2014; 104:47-54. [DOI: 10.1016/j.lfs.2014.03.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 02/13/2014] [Accepted: 03/18/2014] [Indexed: 12/21/2022]
|
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: 1.5] [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
|
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
|
36
|
Boulware MI, Heisler JD, Frick KM. The memory-enhancing effects of hippocampal estrogen receptor activation involve metabotropic glutamate receptor signaling. J Neurosci 2013; 33:15184-94. [PMID: 24048848 PMCID: PMC6618419 DOI: 10.1523/jneurosci.1716-13.2013] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 07/22/2013] [Accepted: 08/10/2013] [Indexed: 01/22/2023] Open
Abstract
Our laboratory has demonstrated that 17β-estradiol (E2) enhances hippocampal memory consolidation via rapid activation of multiple intracellular signaling cascades, including the ERK/MAPK cascade (Fernandez et al., 2008; Fan et al., 2010). However, the receptor mechanisms responsible for these effects of E2 remain unclear. In vitro, estrogen receptor α (ERα) signaling through metabotropic glutamate receptor 1a (mGluR1a) leads to ERK-dependent CREB phosphorylation (Boulware et al., 2005), suggesting that interactions between ERs and mGluR1a may be vital to the memory-enhancing effects of E2. As such, the present study tested the roles of classical estrogen receptors (ERα and ERβ) and mGluR1a in mediating the effects of E2 on hippocampal memory consolidation. Dorsal hippocampal (DH) infusion of ERα (PPT) or ERβ (DPN) agonists enhanced novel object recognition and object placement memory in ovariectomized female mice in an ERK-dependent manner, suggesting that these receptors influence memory by rapidly activating hippocampal cell signaling. Next, DH infusion of the mGluR1a antagonist LY367385 blocked the object and spatial memory facilitation induced by E2, PPT, and DPN, demonstrating that ER/mGluR1a signaling is critical for the memory-enhancing effects of E2. Finally, we show that ERα, ERβ, mGluR1, and ERK all reside within specialized membrane microdomains of the DH, and that ERα and ERβ physically interact with mGluR1, providing a means through which ERs may activate mGluRs and downstream signaling. Together, these findings provide the first in vivo evidence demonstrating that ER/mGluR signaling can mediate the beneficial effects of E2 on hippocampal memory consolidation.
Collapse
Affiliation(s)
- Marissa I. Boulware
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - John D. Heisler
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| | - Karyn M. Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211
| |
Collapse
|
37
|
Lee CWS, Ho IK. Sex differences in opioid analgesia and addiction: interactions among opioid receptors and estrogen receptors. Mol Pain 2013; 9:45. [PMID: 24010861 PMCID: PMC3844594 DOI: 10.1186/1744-8069-9-45] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/03/2013] [Indexed: 12/21/2022] Open
Abstract
Opioids are widely used as the pain reliever and also notorious for being addictive drugs. Sex differences in the opioid analgesia and addiction have been reported and investigated in human subjects and animal models. Yet, the molecular mechanism underlying the differences between males and females is still unclear. Here, we reviewed the literature describing the sex differences in analgesic responses and addiction liabilities to clinically relevant opioids. The reported interactions among opioids, estrogens, opioid receptors, and estrogen receptors are also evaluated. We postulate that the sex differences partly originated from the crosstalk among the estrogen and opioid receptors when stimulated by the exogenous opioids, possibly through common secondary messengers and the downstream gene transcriptional regulators.
Collapse
Affiliation(s)
- Cynthia Wei-Sheng Lee
- Center for Drug Abuse and Addiction, China Medical University Hospital, 2 Yuh-Der Road, Taichung 40447, Taiwan.
| | | |
Collapse
|
38
|
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: 5.7] [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
|
39
|
Borgquist A, Kachani M, Tavitian N, Sinchak K, Wagner EJ. Estradiol negatively modulates the pleiotropic actions of orphanin FQ/nociceptin at proopiomelanocortin synapses. Neuroendocrinology 2013; 98:60-72. [PMID: 23735696 PMCID: PMC4170741 DOI: 10.1159/000351868] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 05/08/2013] [Indexed: 12/11/2022]
Abstract
Orphanin FQ/nociceptin (OFQ/N) inhibits the activity of proopiomelanocortin (POMC) neurons located in the hypothalamic arcuate nucleus (ARH) that regulate female sexual behavior and energy balance. We tested the hypothesis that estradiol modulates the ability of OFQ/N to pre- and postsynaptically decrease the excitability of these cells. To this end, whole-cell patch-clamp recordings were performed in hypothalamic slices prepared from ovariectomized rats, including some that were injected with the retrograde tracer Fluorogold in the medial preoptic nucleus (MPN) to label the POMC neurons regulating sexual receptivity. OFQ/N (1 µM) evoked a robust outward current in ARH neurons from vehicle-treated animals that was blocked by the opioid receptor-like (ORL)1 receptor antagonist UFP-101 (100 nM) and the G protein-gated, inwardly rectifying K⁺ (GIRK-1) channel blocker tertiapin (10 nM). OFQ/N also produced a decrease in the frequency of glutamatergic, miniature excitatory postsynaptic currents (mEPSCs), which was also antagonized by UFP-101. Estradiol benzoate (2 µg) increased basal mEPSC frequency and markedly diminished both the OFQ/N-induced activation of postsynaptic GIRK-1 channel currents and the presynaptic inhibition of glutamatergic neurotransmission. These effects were observed in identified POMC neurons, including eight that projected to the MPN. Taken together, these data reveal that estradiol attenuates the pleiotropic inhibitory actions of OFQ/N on POMC neurons: presynaptically through reducing the OFQ/N inhibition of glutamate release and postsynaptically by reducing ORL1 signaling through GIRK channels. As such, they impart critical insight into a mechanism for estradiol to increase the activity of POMC neurons that inhibit sexual receptivity.
Collapse
Affiliation(s)
- Amanda Borgquist
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
| | - Malika Kachani
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA 91766
| | - Nadia Tavitian
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
| | - Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, Long Beach, CA 90840
| | - Edward J. Wagner
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
40
|
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
|
41
|
Kampa M, Pelekanou V, Notas G, Stathopoulos EN, Castanas E. The estrogen receptor: two or more molecules, multiple variants, diverse localizations, signaling and functions. Are we undergoing a paradigm-shift as regards their significance in breast cancer? Hormones (Athens) 2013; 12:69-85. [PMID: 23624133 DOI: 10.1007/bf03401288] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Marilena Kampa
- Department of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Crete, Greece
| | | | | | | | | |
Collapse
|
42
|
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: 2.5] [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
|
43
|
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: 5.4] [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
|
44
|
Cornil CA, Ball GF, Balthazart J. Rapid control of male typical behaviors by brain-derived estrogens. Front Neuroendocrinol 2012; 33:425-46. [PMID: 22983088 PMCID: PMC3496013 DOI: 10.1016/j.yfrne.2012.08.003] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 08/13/2012] [Accepted: 08/17/2012] [Indexed: 01/01/2023]
Abstract
Beside their genomic mode of action, estrogens also activate a variety of cellular signaling pathways through non-genomic mechanisms. Until recently, little was known regarding the functional significance of such actions in males and the mechanisms that control local estrogen concentration with a spatial and time resolution compatible with these non-genomic actions had rarely been examined. Here, we review evidence that estrogens rapidly modulate a variety of behaviors in male vertebrates. Then, we present in vitro work supporting the existence of a control mechanism of local brain estrogen synthesis by aromatase along with in vivo evidence that rapid changes in aromatase activity also occur in a region-specific manner in response to changes in the social or environmental context. Finally, we suggest that the brain estrogen provision may also play a significant role in females. Together these data bolster the hypothesis that brain-derived estrogens should be considered as neuromodulators.
Collapse
Affiliation(s)
- Charlotte A Cornil
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium.
| | | | | |
Collapse
|
45
|
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: 6.8] [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
|
46
|
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: 1.9] [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
|
47
|
Zhang Y, Xiao X, Zhang XM, Zhao ZQ, Zhang YQ. Estrogen facilitates spinal cord synaptic transmission via membrane-bound estrogen receptors: implications for pain hypersensitivity. J Biol Chem 2012; 287:33268-81. [PMID: 22869379 DOI: 10.1074/jbc.m112.368142] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent evidence suggests that estrogen is synthesized in the spinal dorsal horn and plays a role in nociceptive processes. However, the cellular and molecular mechanisms underlying these effects remain unclear. Using electrophysiological, biochemical, and morphological techniques, we here demonstrate that 17β-estradiol (E2), a major form of estrogen, can directly modulate spinal cord synaptic transmission by 1) enhancing NMDA receptor-mediated synaptic transmission in dorsal horn neurons, 2) increasing glutamate release from primary afferent terminals, 3) increasing dendritic spine density in cultured spinal cord dorsal horn neurons, and 4) potentiating spinal cord long term potentiation (LTP) evoked by high frequency stimulation (HFS) of Lissauer's tract. Notably, E2-BSA, a ligand that acts only on membrane estrogen receptors, can mimic E2-induced facilitation of HFS-LTP, suggesting a nongenomic action of this neurosteroid. Consistently, cell surface biotinylation demonstrated that three types of ERs (ERα, ERβ, and GPER1) are localized on the plasma membrane of dorsal horn neurons. Furthermore, the ERα and ERβ antagonist ICI 182,780 completely abrogates the E2-induced facilitation of LTP. ERβ (but not ERα) activation can recapitulate E2-induced persistent increases in synaptic transmission (NMDA-dependent) and dendritic spine density, indicating a critical role of ERβ in spinal synaptic plasticity. E2 also increases the phosphorylation of ERK, PKA, and NR2B, and spinal HFS-LTP is prevented by blockade of PKA, ERK, or NR2B activation. Finally, HFS increases E2 release in spinal cord slices, which can be prevented by aromatase inhibitor androstatrienedione, suggesting activity-dependent local synthesis and release of endogenous E2.
Collapse
Affiliation(s)
- Yan Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| | | | | | | | | |
Collapse
|
48
|
Fiocchetti M, Ascenzi P, Marino M. Neuroprotective effects of 17β-estradiol rely on estrogen receptor membrane initiated signals. Front Physiol 2012; 3:73. [PMID: 22493583 PMCID: PMC3319910 DOI: 10.3389/fphys.2012.00073] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 03/13/2012] [Indexed: 12/15/2022] Open
Abstract
Besides its crucial role in many physiological events, 17β-estradiol (E2) exerts protective effects in the central nervous system. The E2 effects are not restricted to the brain areas related with the control of reproductive function, but rather are widespread throughout the developing and the adult brain. E2 actions are mediated through estrogen receptors (i.e., ERα and ERβ) belonging to the nuclear receptor super-family. As members of the ligand-regulated transcription factor family, classically, the actions of ERs in the brain were thought to mediate only the E2 long-term transcriptional effects. However, a growing body of evidence highlighted rapid, membrane initiated E2 effects in the brain that are independent of ER transcriptional activities and are involved in E2-induced neuroprotection. The aim of this review is to focus on the rapid effects of E2 in the brain highlighting the specific role of the signaling pathway(s) of the ERβ subtype in the neuroprotective actions of E2.
Collapse
|
49
|
Kelly MJ. Introduction. Neuroendocrinology 2012; 96:101-2. [PMID: 22986409 DOI: 10.1159/000342130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Indexed: 11/19/2022]
Affiliation(s)
- Martin J Kelly
- Department of Physiology and Pharmacology, L334, Oregon Health and Science University, Portland, OR 97239-3098, 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: 9.4] [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
|