Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 17(12): 783-797, 2016 12.
Article in English | MEDLINE | ID: mdl-27729652

ABSTRACT

Steroid hormone receptors mediate numerous crucial biological processes and are classically thought to function as transcriptional regulators in the nucleus. However, it has been known for more than 50 years that steroids evoke rapid responses in many organs that cannot be explained by gene regulation. Mounting evidence indicates that most steroid receptors in fact exist in extranuclear cellular pools, including at the plasma membrane. This latter pool, when engaged by a steroid ligand, rapidly activates signals that affect various aspects of cellular biology. Research into the mechanisms of signalling instigated by extranuclear steroid receptor pools and how this extranuclear signalling is integrated with responses elicited by nuclear receptor pools provides novel understanding of steroid hormone signalling and its roles in health and disease.


Subject(s)
Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Signal Transduction , Animals , Cell Nucleus , Gene Expression Regulation , Humans , Protein Transport
2.
Differentiation ; 118: 24-33, 2021.
Article in English | MEDLINE | ID: mdl-33339644

ABSTRACT

Estrogen signaling through the main estrogen receptor, estrogen receptor 1 (ESR1; also known as ERα), is essential for normal female and male reproductive function. Historically, studies of estrogen action have focused on the classical genomic pathway. Although this is clearly the major pathway for steroid hormone actions, these hormones also signal through rapid non-classical effects involving cell membrane actions. Reports of rapid effects of estrogens extend for more than half a century, but recent results have expanded understanding of the identity, structure, function and overall importance of membrane receptors in estrogen responses. Key findings in this field were the immunohistochemical detection of ESR1 in cell membranes and demonstration that a portion of newly synthesized ESR1 is routed to the membrane by palmitoylation. These receptors in the membrane can then signal through protein kinases and other mechanisms following ligand binding to alter cell function. Another crucial advance in the field was development of transgenic mice expressing normal amounts of functional nuclear ESR1 (nESR1) but lacking membrane ESR1 (mESR1). Both male and female transgenic mice lacking mESR1 were infertile as adults, and both sexes had extensive reproductive abnormalities. Transgenic mice lacking mESR1 were highly protected from deleterious effects of neonatal estrogen administration, and estrogen effects on the histone methyltransferase Enhancer of Zeste homolog 2 that are mediated through mESR1 could have significant effects on epigenetic imprinting. In summary, signaling through mESR1 is essential for normal male and female reproductive function and fertility, and is a critical enabler of normal estrogen responses in vivo. Although the precise role of mESR1 in estrogen responses remains to be established, future research in this area should clarify its mechanism of action and lead to a better understanding of how mESR1 signaling works with classical genomic signaling through nESR1 to promote full estrogenic responses.


Subject(s)
Cell Nucleus/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Estrogen Receptor alpha/genetics , Genitalia/metabolism , Animals , Cell Membrane/genetics , Epigenesis, Genetic/genetics , Female , Genitalia/physiology , Genitalia, Female/metabolism , Genitalia, Female/physiology , Genitalia, Male/metabolism , Genitalia, Male/physiology , Genomic Imprinting/genetics , Humans , Male , Mice, Transgenic/genetics , Signal Transduction/genetics
3.
J Neurosci ; 38(37): 7935-7951, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209204

ABSTRACT

Men are generally superior to women in remembering spatial relationships, whereas the reverse holds for semantic information, but the neurobiological bases for these differences are not understood. Here we describe striking sexual dimorphism in synaptic mechanisms of memory encoding in hippocampal field CA1, a region critical for spatial learning. Studies of acute hippocampal slices from adult rats and mice show that for excitatory Schaffer-commissural projections, the memory-related long-term potentiation (LTP) effect depends upon endogenous estrogen and membrane estrogen receptor α (ERα) in females but not in males; there was no evident involvement of nuclear ERα in females, or of ERß or GPER1 (G-protein-coupled estrogen receptor 1) in either sex. Quantitative immunofluorescence showed that stimulation-induced activation of two LTP-related kinases (Src, ERK1/2), and of postsynaptic TrkB, required ERα in females only, and that postsynaptic ERα levels are higher in females than in males. Several downstream signaling events involved in LTP were comparable between the sexes. In contrast to endogenous estrogen effects, infused estradiol facilitated LTP and synaptic signaling in females via both ERα and ERß. The estrogen dependence of LTP in females was associated with a higher threshold for both inducing potentiation and acquiring spatial information. These results indicate that the observed sexual dimorphism in hippocampal LTP reflects differences in synaptic kinase activation, including both a weaker association with NMDA receptors and a greater ERα-mediated kinase activation in response to locally produced estrogen in females. We propose that male/female differences in mechanisms and threshold for field CA1 LTP contribute to differences in encoding specific types of memories.SIGNIFICANCE STATEMENT There is good evidence for male/female differences in memory-related cognitive function, but the neurobiological basis for this sexual dimorphism is not understood. Here we describe sex differences in synaptic function in a brain area that is critical for learning spatial cues. Our results show that female rodents have higher synaptic levels of estrogen receptor α (ERα) and, in contrast to males, require membrane ERα for the activation of signaling kinases that support long-term potentiation (LTP), a form of synaptic plasticity thought to underlie learning. The additional requirement of estrogen signaling in females resulted in a higher threshold for both LTP and hippocampal field CA1-dependent spatial learning. These results describe a synaptic basis for sexual dimorphism in encoding spatial information.


Subject(s)
Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Sex Characteristics , Spatial Learning/physiology , Synapses/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Enzyme Inhibitors/pharmacology , Estradiol/pharmacology , Estrogen Receptor Modulators/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/drug effects , Male , Memory/drug effects , Mice , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/physiology , Phosphorylation , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Spatial Learning/drug effects , Synapses/drug effects , rho-Associated Kinases/antagonists & inhibitors
4.
Biol Reprod ; 101(2): 392-404, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31141131

ABSTRACT

Both membrane and nuclear fractions of estrogen receptor 1 (ESR1) mediate 17ß-estradiol (E2) actions. Mice expressing nuclear (n)ESR1 but lacking membrane (m)ESR1 (nuclear-only estrogen receptor 1 [NOER] mice) show reduced E2 responsivity and reproductive abnormalities culminating in adult male and female infertility. Using this model, we investigated whether reproductive pathologies caused by the synthetic estrogen diethylstilbestrol (DES) are mitigated by mESR1 ablation. Homozygous and heterozygous wild-type (WT and HET, respectively) and NOER male and female mice were subcutaneously injected with DES (1 mg/kg body weight [BW]) or vehicle daily from postnatal day (PND) 1-5. Uterine histology was assessed in select DES-treated females at PND 5, whereas others were ovariectomized at PND 60 and treated with E2 (10 µg/kg BW) or vehicle 2 weeks later. Neonatal DES exposure resulted in ovary-independent epithelial proliferation in the vagina and uterus of WT but not NOER females. Neonatal DES treatment also induced ovary-independent adult expression of classical E2-induced transcripts (e.g., lactoferrin [Ltf] and enhancer of zeste homolog 2 [Ezh2]) in WT but not NOER mice. At PND 90, DES-treated WT and HET males showed smaller testes and a high incidence of bacterial pyogranulomatous inflammation encompassing the testes, epididymis and occasionally the ductus deferens with spread to lumbar lymph nodes; such changes were largely absent in NOER males. Results indicate that male and female NOER mice are protected from deleterious effects of neonatal DES, and thus mESR1 signaling is required for adult manifestation of DES-induced reproductive pathologies in both sexes.


Subject(s)
Diethylstilbestrol/toxicity , Estrogen Receptor alpha/genetics , Estrogens, Non-Steroidal/toxicity , Prenatal Exposure Delayed Effects , Animals , Female , Gene Expression Regulation/drug effects , Genital Diseases, Male/chemically induced , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Uterus/metabolism
5.
Biol Reprod ; 101(2): 306-317, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31201420

ABSTRACT

Enhancer of zeste homolog 2 (EZH2) is a rate-limiting catalytic subunit of a histone methyltransferase, polycomb repressive complex, which silences gene activity through the repressive histone mark H3K27me3. EZH2 is critical for epigenetic effects of early estrogen treatment, and may be involved in uterine development and pathologies. We investigated EZH2 expression, regulation, and its role in uterine development/function. Uterine epithelial EZH2 expression was associated with proliferation and was high neonatally then declined by weaning. Pre-weaning uterine EZH2 expression was comparable in wild-type and estrogen receptor 1 knockout mice, showing neonatal EZH2 expression is ESR1 independent. Epithelial EZH2 was upregulated by 17ß-estradiol (E2) and inhibited by progesterone in adult uteri from ovariectomized mice. To investigate the uterine role of EZH2, we developed a EZH2 conditional knockout (Ezh2cKO) mouse using a cre recombinase driven by the progesterone receptor (Pgr) promoter that produced Ezh2cKO mice lacking EZH2 in Pgr-expressing tissues (e.g. uterus, mammary glands). In Ezh2cKO uteri, EZH2 was deleted neonatally. These uteri had reduced H3K27me3, were larger than WT, and showed adult cystic endometrial hyperplasia. Ovary-independent uterine epithelial proliferation and increased numbers of highly proliferative uterine glands were seen in adult Ezh2cKO mice. Female Ezh2cKO mice were initially subfertile, and then became infertile by 9 months. Mammary gland development in Ezh2cKO mice was inhibited. In summary, uterine EZH2 expression is developmentally and hormonally regulated, and its loss causes aberrant uterine epithelial proliferation, uterine hypertrophy, and cystic endometrial hyperplasia, indicating a critical role in uterine development and function.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Developmental/physiology , Gene Expression Regulation, Enzymologic/physiology , Uterus/enzymology , Uterus/growth & development , Animals , Enhancer of Zeste Homolog 2 Protein/genetics , Epithelial Cells/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Female , Histones/metabolism , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice , Mice, Knockout , Pregnancy , Progesterone/metabolism
6.
Annu Rev Med ; 66: 271-80, 2015.
Article in English | MEDLINE | ID: mdl-25587652

ABSTRACT

Steroid hormones are produced throughout the phylogenetic tree, from plants to mammals. In the past 40 years, steroid receptors localized to the nucleus have been recognized as being important to mediating steroid action in many organs. This action mainly arises from the regulation of key genes that are important for organ development and function. These include but are not limited to genes influencing the reproductive tract, mammary glands, bone, brain, fat differentiation, pituitary hormone regulation, and metabolic effects in many organs. Unfortunately, steroids also promote the development of hormone-responsive cancers, including breast, uterus, and prostate cancer. It has also been shown that steroid receptors exist outside the nucleus in many organs and cells, with unclear impact for normal development, health, and disease. This review describes the evidence from many laboratories that these receptors exist and function with nuclear receptors to provide the full impact of all steroid hormones.


Subject(s)
Breast Neoplasms/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Estrogens/metabolism , Mitochondria/metabolism , Receptors, Estrogen/metabolism , Female , Gonadal Steroid Hormones/metabolism , Humans , Receptors, Steroid/metabolism , Signal Transduction
7.
Am J Physiol Endocrinol Metab ; 307(2): E133-40, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24895281

ABSTRACT

Steroid receptors exist and function in multiple compartments of cells in most organs. Although the functions and nature of some of these receptors is being defined, important aspects of receptor localization and signaling to physiology and pathophysiology have been identified. In particular, extranuclear sex steroid receptors have been found in many normal cells and in epithelial tumors, where they enact signal transduction that impacts both nongenomic and genomic functions. Here, I focus on the progress made in understanding the roles of extranuclear estrogen receptors (ER) in physiology and pathophysiology. Extranuclear ER serve as a model to selectively intervene with novel receptor reagents to prevent or limit disease progression. Recent novel mouse models and membrane ER-selective agonists also provide a better understanding of receptor pool cross-talk that results in the overall integrative actions of sex steroids.


Subject(s)
Cell Membrane/metabolism , Receptors, Estrogen/physiology , Animal Structures/physiology , Animals , Humans , Mice , Models, Animal , Organogenesis/genetics , Protein Transport
8.
Biology (Basel) ; 12(4)2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37106832

ABSTRACT

Estrogens via estrogen receptor alpha (ERα) genomic and nongenomic signaling can influence plasticity processes in numerous brain regions. Using mice that express nuclear only ERα (NOER) or membrane only ERα (MOER), this study examined the effect of receptor compartmentalization on the paraventricular nucleus of the hypothalamus (PVN) and the hippocampus. The absence of nuclear and membrane ERα expression impacted females but not males in these two brain areas. In the PVN, quantitative immunohistochemistry showed that the absence of nuclear ERα increased nuclear ERß. Moreover, in the hippocampus CA1, immuno-electron microscopy revealed that the absence of either nuclear or membrane ERα decreased extranuclear ERα and pTrkB in synapses. In contrast, in the dentate gyrus, the absence of nuclear ERα increased pTrkB in synapses, whereas the absence of membrane ERα decreased pTrkB in axons. However, the absence of membrane only ERα decreased the sprouting of mossy fibers in CA3 as reflected by changes in zinc transporter immunolabeling. Altogether these findings support the idea that both membrane and nuclear ERα contribute overlapping and unique actions of estrogen that are tissue- and cellular-specific.

9.
Endocr Rev ; 43(4): 720-742, 2022 07 13.
Article in English | MEDLINE | ID: mdl-34791092

ABSTRACT

Rapid effects of steroid hormones were discovered in the early 1950s, but the subject was dominated in the 1970s by discoveries of estradiol and progesterone stimulating protein synthesis. This led to the paradigm that steroid hormones regulate growth, differentiation, and metabolism via binding a receptor in the nucleus. It took 30 years to appreciate not only that some cellular functions arise solely from membrane-localized steroid receptor (SR) actions, but that rapid sex steroid signaling from membrane-localized SRs is a prerequisite for the phosphorylation, nuclear import, and potentiation of the transcriptional activity of nuclear SR counterparts. Here, we provide a review and update on the current state of knowledge of membrane-initiated estrogen (ER), androgen (AR) and progesterone (PR) receptor signaling, the mechanisms of membrane-associated SR potentiation of their nuclear SR homologues, and the importance of this membrane-nuclear SR collaboration in physiology and disease. We also highlight potential clinical implications of pathway-selective modulation of membrane-associated SR.


Subject(s)
Receptors, Progesterone , Receptors, Steroid , Androgens , Estradiol , Estrogens , Humans , Progesterone/physiology , Receptors, Androgen , Receptors, Progesterone/metabolism , Receptors, Steroid/metabolism , Steroids
10.
Endocrinology ; 164(2)2022 12 19.
Article in English | MEDLINE | ID: mdl-36461668

ABSTRACT

Estrogen acting through estrogen receptor ß (ERß) has been shown to oppose the stimulation of cardiac myocytes and cardiac fibroblasts that results in cardiac hypertrophy and fibrosis. Previous work has implicated signal transduction from ERß as being important to the function of estrogen in this regard. Here we address whether membrane ERß is sufficient to oppose key mechanisms by which angiotensin II (AngII) stimulates cardiac cell pathology. To do this we first defined essential structural elements within ERß that are necessary for membrane or nuclear localization in cells. We previously determined that cysteine 418 is the site of palmitoylation of ERß that is required and sufficient for cell membrane localization in mice and is the same site in humans. Here we determined in Chinese hamster ovarian (CHO) cells, and mouse and rat myocytes and cardiac fibroblasts, the effect on multiple aspects of signal transduction by expressing wild-type (WT ) or a C418A-mutant ERß. To test the importance of the nuclear receptor, we determined a 4-amino acid deletion in the E domain of ERß that strongly blocked nuclear localization. Using these tools, we expressed WT and mutant ERß constructs into cardiomyocytes and cardiac fibroblasts from ERß-deleted mice. We determined the ability of estrogen to mitigate cell pathology stimulated by AngII and whether the membrane ERß is necessary and sufficient.


Subject(s)
Cardiomegaly , Estrogen Receptor beta , Myocytes, Cardiac , Animals , Cricetinae , Mice , Rats , Angiotensin II/pharmacology , Angiotensin II/metabolism , Cardiomegaly/metabolism , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Estrogens/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
11.
JAMA Netw Open ; 5(4): e227852, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35438754

ABSTRACT

Importance: SARS-CoV-2 entry requires the TMPRSS2 cell surface protease. Antiandrogen therapies reduce expression of TMPRSS2. Objective: To determine if temporary androgen suppression induced by degarelix improves clinical outcomes of inpatients hospitalized with COVID-19. Design, Setting, and Participants: The Hormonal Intervention for the Treatment in Veterans With COVID-19 Requiring Hospitalization (HITCH) phase 2, placebo-controlled, double-blind, randomized clinical trial compared efficacy of degarelix plus standard care vs placebo plus standard care on clinical outcomes in men hospitalized with COVID-19 but not requiring invasive mechanical ventilation. Inpatients were enrolled at 14 Department of Veterans Affairs hospitals from July 22, 2020, to April 8, 2021. Data were analyzed from August 9 to October 15, 2021. Interventions: Patients stratified by age, history of hypertension, and disease severity were centrally randomized 2:1 to degarelix, (1-time subcutaneous dose of 240 mg) or a saline placebo. Standard care included but was not limited to supplemental oxygen, antibiotics, vasopressor support, peritoneal dialysis or hemodialysis, intravenous fluids, remdesivir, convalescent plasma, and dexamethasone. Main Outcomes and Measures: The composite primary end point was mortality, ongoing need for hospitalization, or requirement for mechanical ventilation at day 15 after randomization. Secondary end points were time to clinical improvement, inpatient mortality, length of hospitalization, duration of mechanical ventilation, time to achieve a temperature within reference range, maximum severity of COVID-19, and the composite end point at 30 days. Results: The trial was stopped for futility after the planned interim analysis, at which time there were 96 evaluable patients, including 62 patients randomized to the degarelix group and 34 patients in the placebo group, out of 198 initially planned. The median (range) age was 70.5 (48-85) years. Common comorbidities included chronic obstructive pulmonary disorder (15 patients [15.6%]), hypertension (75 patients [78.1%]), cardiovascular disease (27 patients [28.1%]), asthma (12 patients [12.5%]), diabetes (49 patients [51.0%]), and chronic respiratory failure requiring supplemental oxygen at baseline prior to COVID-19 (9 patients [9.4%]). For the primary end point, there was no significant difference between the degarelix and placebo groups (19 patients [30.6%] vs 9 patients [26.5%]; P = .67). Similarly, no differences were observed between degarelix and placebo groups in any secondary end points, including inpatient mortality (11 patients [17.7%] vs 6 patients [17.6%]) or all-cause mortality (11 patients [17.7%] vs 7 patents [20.6%]). There were no differences between degarelix and placebo groups in the overall rates of adverse events (13 patients [21.0%] vs 8 patients [23.5%) and serious adverse events (19 patients [30.6%] vs 13 patients [32.4%]), nor unexpected safety concerns. Conclusions and Relevance: In this randomized clinical trial of androgen suppression vs placebo and usual care for men hospitalized with COVID-19, degarelix did not result in amelioration of COVID-19 severity. Trial Registration: ClinicalTrials.gov Identifier: NCT04397718.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Hypertension , Aged , Aged, 80 and over , Androgens , COVID-19/therapy , Hospitalization , Humans , Immunization, Passive , Male , Oxygen , SARS-CoV-2 , Treatment Outcome , United States , COVID-19 Serotherapy
12.
Endocr Rev ; 28(7): 726-41, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17916740

ABSTRACT

Rapid effects of steroid hormones result from the actions of specific receptors localized most often to the plasma membrane. Fast-acting membrane-initiated steroid signaling (MISS) leads to the modification of existing proteins and cell behaviors. Rapid steroid-triggered signaling through calcium, amine release, and kinase activation also impacts the regulation of gene expression by steroids, sometimes requiring integration with nuclear steroid receptor function. In this and other ways, the integration of all steroid actions in the cell coordinates outcomes such as cell fate, proliferation, differentiation, and migration. The nature of the receptors is of intense interest, and significant data suggest that extranuclear and nuclear steroid receptor pools are the same proteins. Insights regarding the structural determinants for membrane localization and function, as well as the nature of interactions with G proteins and other signaling molecules in confined areas of the membrane, have led to a fuller understanding of how steroid receptors effect rapid actions. Increasingly, the relevance of rapid signaling for the in vivo functions of steroid hormones has been established. Examples include steroid effects on reproductive organ development and function, cardiovascular responsiveness, and cancer biology. However, although great strides have been made, much remains to be understood concerning the integration of extranuclear and nuclear receptor functions to organ biology. In this review, we highlight the significant progress that has been made in these areas.


Subject(s)
Receptors, Steroid/metabolism , Animals , Humans , Signal Transduction
13.
Endocrinology ; 161(9)2020 09 01.
Article in English | MEDLINE | ID: mdl-32730568

ABSTRACT

Severe outcomes and death from the novel coronavirus disease 2019 (COVID-19) appear to be characterized by an exaggerated immune response with hypercytokinemia leading to inflammatory infiltration of the lungs and acute respiratory distress syndrome. Risk of severe COVID-19 outcomes is consistently lower in women than men worldwide, suggesting that female biological sex is instrumental in protection. This mini-review discusses the immunomodulatory and anti-inflammatory actions of high physiological concentrations of the steroids 17ß-estradiol (E2) and progesterone (P4). We review how E2 and P4 favor a state of decreased innate immune inflammatory response while enhancing immune tolerance and antibody production. We discuss how the combination of E2 and P4 may improve the immune dysregulation that leads to the COVID-19 cytokine storm. It is intended to stimulate novel consideration of the biological forces that are protective in women compared to men, and to therapeutically harness these factors to mitigate COVID-19 morbidity and mortality.


Subject(s)
Coronavirus Infections/immunology , Estradiol/immunology , Immunomodulation/immunology , Pneumonia, Viral/immunology , Progesterone/immunology , Antibody Formation/immunology , Betacoronavirus , COVID-19 , Contraceptives, Oral, Hormonal/therapeutic use , Coronavirus Infections/drug therapy , Coronavirus Infections/mortality , Coronavirus Infections/physiopathology , Cytokine Release Syndrome/immunology , Drug Repositioning , Estradiol/therapeutic use , Estrogen Replacement Therapy , Estrogens/therapeutic use , Female , Humans , Immune Tolerance/immunology , Immunity, Innate/immunology , Male , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/mortality , Pneumonia, Viral/physiopathology , Pregnancy , Pregnancy Complications, Infectious/immunology , Progesterone/therapeutic use , Progestins/therapeutic use , SARS-CoV-2 , Selective Estrogen Receptor Modulators/therapeutic use , Severity of Illness Index , Sex Factors , COVID-19 Drug Treatment
14.
Endocrinology ; 161(11)2020 11 01.
Article in English | MEDLINE | ID: mdl-32976570

ABSTRACT

Mesenchymal stem cells can differentiate into mature chondrocytes, osteoblasts, and adipocytes. Excessive and dysfunctional visceral adipocytes increase upon menopause and importantly contribute to altered metabolism in postmenopausal women. We previously showed both plasma membrane and nuclear estrogen receptors alpha (ERα) with endogenous estrogen are required to suppress adipogenesis in vivo. Here we determined mechanisms by which these liganded ER pools collaborate to inhibit the peroxisome proliferator-activated gamma (PPARγ) gene and subsequent progenitor differentiation. In 3T3-L1 pre-adipocytes and adipose-derived stem cells (ADSC), membrane ERα signaled through phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT) to enhance ERα nuclear localization, importantly at the PPARγ gene promoter. AKT also increased overall abundance and recruitment of co-repressors GATA3, ß-catenin, and TCF4 to the PPARγ promoter. Membrane ERα signaling additionally enhanced wingless-integrated (Wnt)1 and 10b expression. The components of the repressor complex were required for estrogen to inhibit rosiglitazone-induced differentiation of ADSC and 3T3-L1 cells to mature adipocytes. These mechanisms whereby ER cellular pools collaborate to inhibit gene expression limit progenitor differentiation to mature adipocytes.


Subject(s)
Adipogenesis/genetics , Estrogen Receptor alpha/physiology , 3T3-L1 Cells , Adipocytes/physiology , Animals , Cell Differentiation/genetics , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Down-Regulation/genetics , Estrogen Receptor alpha/metabolism , Female , Mice , Mice, Transgenic , Signal Transduction/genetics
15.
J Physiol ; 587(Pt 21): 5019-23, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19687123

ABSTRACT

Conservation of steroid hormone action outside the nucleus occurs from plants that make brassinosteroids to higher metazoans (primates). In plants, steroid hormone action occurs when the brassinosteroids bind a membrane tyrosine kinase receptor. Ligated receptors for all sex steroids exist at the plasma membrane and rapidly signal through G proteins to second messengers including calcium, cAMP and cGMP, activating proximal and more distal kinases. These signal cascades impact many functions of steroid hormones, responsible for the biological actions of these molecules. Support also exists for membrane-localized receptors of other members of the steroid superfamily, responding to glucocorticoids, mineralocorticoids, thyroid hormone, and vitamin D. The nature of these receptors is in some cases unclear. Steroid receptors also exist in discrete cytoplasmic organelles, most notably the mitochondria, although the functions of these receptors are poorly understood. In this review, I highlight the essential elements of the membrane oestrogen receptor alpha, noting where conserved aspects exist for other steroid receptors.


Subject(s)
Cell Membrane/physiology , Estrogen Receptor alpha/metabolism , Models, Biological , Signal Transduction/physiology , Animals , Humans
16.
Mol Biol Cell ; 17(5): 2125-37, 2006 May.
Article in English | MEDLINE | ID: mdl-16495339

ABSTRACT

Steroid hormones have been reported to indirectly impact mitochondrial functions, attributed to nuclear receptor-induced production of proteins that localize in this cytoplasmic organelle. Here we show high-affinity estrogen receptors in the mitochondria of MCF-7 breast cancer cells and endothelial cells, compatible with classical estrogen receptors ERalpha and ERbeta. We report that in MCF-7, estrogen inhibits UV radiation-induced cytochrome C release, the decrease of the mitochondrial membrane potential, and apoptotic cell death. UV stimulated the formation of mitochondrial reactive oxygen species (mROS), and mROS were essential to inducing mitochondrial events of cell death. mROS mediated the UV activation of c-jun N-terminal kinase (JNK), and protein kinase C (PKC) delta, underlying the subsequent translocation of Bax to the mitochondria where oligomerization was promoted. E2 (estradiol) inhibited all these events, directly acting in mitochondria to inhibit mROS by rapidly up-regulating manganese superoxide dismutase activity. We implicate novel functions of ER in the mitochondria of breast cancer that lead to the survival of the tumor cells.


Subject(s)
Breast Neoplasms/metabolism , Mitochondria/chemistry , Mitochondria/metabolism , Receptors, Estrogen/analysis , Receptors, Estrogen/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cytochromes c/metabolism , Estradiol/pharmacology , Estrogens/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/antagonists & inhibitors , bcl-2-Associated X Protein/metabolism
17.
J Clin Invest ; 129(5): 1818-1826, 2019 05 01.
Article in English | MEDLINE | ID: mdl-31042159

ABSTRACT

Androgens and estrogens are known to be critical regulators of mammalian physiology and development. While these two classes of steroids share similar structures (in general, estrogens are derived from androgens via the enzyme aromatase), they subserve markedly different functions via their specific receptors. In the past, estrogens such as estradiol were thought to be most important in the regulation of female biology, while androgens such as testosterone and dihydrotestosterone were believed to primarily modulate development and physiology in males. However, the emergence of patients with deficiencies in androgen or estrogen hormone synthesis or actions, as well as the development of animal models that specifically target androgen- or estrogen-mediated signaling pathways, have revealed that estrogens and androgens regulate critical biological and pathological processes in both males and females. In fact, the concept of "male" and "female" hormones is an oversimplification of a complex developmental and biological network of steroid actions that directly impacts many organs. In this Review, we will discuss important roles of estrogens in males and androgens in females.


Subject(s)
Androgens/physiology , Estrogens/physiology , Animals , Bone and Bones/physiology , Breast Neoplasms/pathology , Central Nervous System/physiology , Dihydrotestosterone , Disease Progression , Estradiol/physiology , Female , Genitalia/physiology , Humans , Male , Mice , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Receptors, Estrogen/physiology , Sex Factors , Testosterone/physiology
18.
Mol Metab ; 22: 62-70, 2019 04.
Article in English | MEDLINE | ID: mdl-30797705

ABSTRACT

OBJECTIVE: The endogenous estrogen 17ß-estradiol (E2) promotes metabolic homeostasis in premenopausal women. In a mouse model of post-menopausal metabolic syndrome, we reported that estrogens increased energy expenditure, thus preventing estrogen deficiency-induced adiposity. Estrogens' prevention of fat accumulation was associated with increased serum concentrations of fibroblast growth factor 21 (FGF21), suggesting that FGF21 participates in estrogens' promotion of energy expenditure. METHODS: We studied the effect of E2 on FGF21 production and the role of FGF21 in E2 stimulation of energy expenditure and prevention of adiposity, using female estrogen receptor (ER)- and FGF21-deficient mice fed a normal chow and a cohort of ovariectomized women from the French E3N prospective cohort study. RESULTS: E2 acting on the hepatocyte ERα increases hepatic expression and production of FGF21 in female mice. In vivo activation of ERα increases the transcription of Fgf21 via an estrogen response element outside the promoter of Fgf21. Treatment with E2 increases oxygen consumption and energy expenditure and prevents whole body fat accumulation in ovariectomized female WT mice. The effect of E2 on energy expenditure is not observed in FGF21-deficient mice. While E2 treatment still prevents fat accumulation in FGF21-deficient mice, this effect is decreased compared to WT mice. In an observational cohort of ovariectomized women, E2 treatment was associated with lower serum FGF21 concentrations, which may reflect a healthier metabolic profile. CONCLUSIONS: In female mice, E2 action on the hepatocyte ERα increases Fgf21 transcription and FGF21 production, thus promoting energy expenditure and partially decreasing fat accumulation.


Subject(s)
Estrogen Receptor alpha/metabolism , Fibroblast Growth Factors/biosynthesis , Animals , Energy Metabolism , Female , Fibroblast Growth Factors/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Diabetes ; 68(3): 490-501, 2019 03.
Article in English | MEDLINE | ID: mdl-30305367

ABSTRACT

Estrogens favor glucose homeostasis primarily through the estrogen receptor-α (ERα), but the respective importance of nuclear ERα (NOER) and membrane ERα (MOER) pools to glucose homeostasis are unknown. We studied glucose homeostasis, insulin secretion, and insulin sensitivity in male and female mice expressing either the NOER or the MOER. Male and female MOER mice exhibited fasting and fed hyperglycemia and glucose intolerance. Female MOER mice displayed impaired central insulin signaling associated with hyperinsulinemia and insulin resistance due to unrestrained hepatic gluconeogenesis, without alterations in glucose-stimulated insulin secretion (GSIS). In contrast, male MOER mice did not exhibit detectable insulin resistance, but showed impaired GSIS associated with reduced brain glucose sensing. Female NOER mice exhibited milder hepatic insulin resistance and glucose intolerance. In conclusion, nuclear ERα signaling is predominant in maintaining glucose homeostasis in mice of both sexes. Lack of nuclear ERα alters the central control of insulin sensitivity in females and predominantly impairs the central regulation of insulin secretion in males.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Estrogen Receptor alpha/metabolism , Receptors, Estrogen/metabolism , Animals , Blood Glucose/metabolism , Female , Immunohistochemistry , Insulin/blood , Insulin Resistance/physiology , Insulin Secretion/physiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL
20.
Endocrinology ; 149(7): 3361-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18372323

ABSTRACT

Estrogen has been reported to prevent development of cardiac hypertrophy in female rodent models and in humans. However, the mechanisms of sex steroid action are incompletely understood. We determined the cellular effects by which 17beta-estradiol (E2) inhibits angiotensin II (AngII)-induced cardiac hypertrophy in vivo. Two weeks of angiotensin infusion in female mice resulted in marked hypertrophy of the left ventricle, exacerbated by the loss of ovarian steroid hormones from oophorectomy. Hypertrophy was 51% reversed by the administration of E2 (insertion of 0.1 mg/21-d-release tablets). The effects of E2 were mainly mediated by the estrogen receptor (ER) beta-isoform, because E2 had little effect in ERbeta-null mice but comparably inhibited AngII-induced hypertrophy in wild-type or ERalpha-null mice. AngII induced a switch of myosin heavy chain production from alpha to beta, but this was inhibited by E2 via ERbeta. AngII-induced ERK activation was also inhibited by E2 through the beta-receptor. E2 stimulated brain natriuretic peptide protein expression and substantially prevented ventricular interstitial cardiac fibrosis (collagen deposition) as induced by AngII. Importantly, E2 inhibited calcineurin activity that was stimulated by AngII, related to E2 stimulating the modulatory calcineurin-interacting protein (MCIP) 1 gene and protein expression. E2 acting mainly through ERbeta mitigates the important signaling by AngII that produces cardiac hypertrophy and fibrosis in female mice.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/drug therapy , Estrogen Receptor beta/physiology , Estrogens/pharmacology , Angiotensin II/pharmacology , Animals , Calcium-Binding Proteins , Cardiomegaly/chemically induced , Collagen/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fibrosis , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myocardium/metabolism , Myocardium/pathology , Myosin Heavy Chains/metabolism , Ovariectomy
SELECTION OF CITATIONS
SEARCH DETAIL