1
|
Cai M, McNamara K, Yamazaki Y, Harada N, Miyashita M, Tada H, Ishida T, Sasano H. The role of mineralocorticoids and glucocorticoids under the impact of 11β-hydroxysteroid dehydrogenase in human breast lesions. Med Mol Morphol 2022; 55:110-122. [PMID: 35103835 DOI: 10.1007/s00795-022-00312-1] [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: 10/25/2021] [Accepted: 01/09/2022] [Indexed: 12/24/2022]
Abstract
We attempted to explore the possible involvement of the in situ availability of mineralocorticoids and mineralocorticoid receptor (MR) in the pathogenesis of mammary ductal carcinoma. We also explored their individual profiles among different subtypes of invasive ductal carcinomas of no special type (IDC-NST) by evaluating the status of MR, Glucocorticoid receptor (GR), and 11β hydroxysteroid dehydrogenase (HSD) 1/2 at each stage of the putative cascade of the mammary ductal proliferative disorders. In this study, IDC-NST, ductal carcinoma in situ (DCIS), atypical ductal hyperplasia (ADH), and non-pathological breast tissues were all evaluated by immunohistochemistry. MR was significantly lower in ADH than in DCIS or IDC-NST. 11βHSD2 was significantly lower in ADH than normal breast tissue and 11βHSD1 was significantly higher in DCIS than normal, ADH, or IDC-NST. MR in progesterone receptor (PR)-positive IDC-NST cases tended to be associated with the Ki-67 labeling index. Results of the present study demonstrated that the status of MR and GR in conjunction with the 11βHSDs was correlated with the development of low-grade proliferative disorders in mammary glands. In addition, the potential crosstalk between MR and PR could also influence cell proliferation of breast carcinoma cells but further investigations are required for clarification.
Collapse
Affiliation(s)
- Mingzhen Cai
- Department of Breast and Endocrine Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Keely McNamara
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuto Yamazaki
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Narumi Harada
- Department of Breast and Endocrine Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Hiroshi Tada
- Department of Breast and Endocrine Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Takanori Ishida
- Department of Breast and Endocrine Surgery, Tohoku University School of Medicine, Sendai, Japan
| | - Hironobu Sasano
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan. .,Department of Pathology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-Ku, Sendai, 980-8575, Japan.
| |
Collapse
|
2
|
Grossmann C, Almeida-Prieto B, Nolze A, Alvarez de la Rosa D. Structural and molecular determinants of mineralocorticoid receptor signalling. Br J Pharmacol 2021; 179:3103-3118. [PMID: 34811739 DOI: 10.1111/bph.15746] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/19/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022] Open
Abstract
During the past decades, the mineralocorticoid receptor (MR) has evolved from a much-overlooked member of the steroid hormone receptor family to an important player, not only in volume and electrolyte homeostasis but also in pathological changes occurring in an increasing number of tissues, especially the renal and cardiovascular systems. Simultaneously, a wealth of information about the structure, interaction partners and chromatin requirements for genomic signalling of steroid hormone receptors became available. However, much of the information for the MR has been deduced from studies of other family members and there is still a lack of knowledge about MR-specific features in ligand binding, chromatin remodelling, co-factor interactions and general MR specificity-conferring mechanisms that can completely explain the differences in pathophysiological function between MR and its closest relative, the glucocorticoid receptor. This review aims to give an overview of the current knowledge of MR structure, signalling and co-factors modulating its activity.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle, Saale, Germany
| | - Brian Almeida-Prieto
- Departamento de Ciencias Médicas Básicas and Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| | - Alexander Nolze
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle, Saale, Germany
| | - Diego Alvarez de la Rosa
- Departamento de Ciencias Médicas Básicas and Instituto de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, Tenerife, Spain
| |
Collapse
|
3
|
Takedomi K, Ohta M, Ekimoto T, Ikeguchi M. Effect of Water Molecules on the Activating S810L Mutation of the Mineralocorticoid Receptor. J Chem Inf Model 2021; 61:3583-3592. [PMID: 34228431 DOI: 10.1021/acs.jcim.1c00389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mineralocorticoid receptor (MR) is a nuclear receptor whose endogenous ligands are mineralocorticoids, a type of steroid hormone. The activating S810L mutation is known to cause severe early-onset and pregnancy-related hypertension. Progesterone binds to the wild-type (WT) MR as a passive antagonist with fast dissociation; however, it binds to the S810L mutant as a full agonist with slow dissociation. The switch in the biological activity of progesterone is considered to be one of the causes of the disease. First, we used steered molecular dynamics simulations to analyze the dissociation process of progesterone for the WT and the S810L mutant. Progesterone in the WT dissociated from the ligand-binding pocket with a weak force in comparison with progesterone in the S810L mutant due to the large inflow of water molecules into the pocket. Therefore, we used conventional molecular dynamics simulations for the ligand-free structures of the WT and the S810L mutant to investigate the effect of the mutation on the inflow of water. In the WT, water molecules enter the ligand-binding pocket in two ways: in the vicinity of (i) Arg817 and (ii) Ser810. In contrast, few water molecules enter the pocket in the S810L mutant because of the large size and hydrophobic nature of the Leu810 side chain. Fast dissociation is a common feature among passive antagonists of MR; therefore, we inferred that the water inflow could be responsible for the dissociation kinetics of progesterone in the WT and the S810L mutant.
Collapse
Affiliation(s)
- Kei Takedomi
- Graduate School of Medicinal Life Science, Yokohama City University, Yokohama 230-0045, Japan.,Modality Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida, Aoba-ku, Yokohama 227-0033, Japan
| | - Masateru Ohta
- HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, Yokohama 230-0045, Japan
| | - Toru Ekimoto
- Graduate School of Medicinal Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Mitsunori Ikeguchi
- Graduate School of Medicinal Life Science, Yokohama City University, Yokohama 230-0045, Japan.,HPC- and AI-driven Drug Development Platform Division, Center for Computational Science, RIKEN, Yokohama 230-0045, Japan
| |
Collapse
|
4
|
Floriou-Servou A, von Ziegler L, Waag R, Schläppi C, Germain PL, Bohacek J. The Acute Stress Response in the Multiomic Era. Biol Psychiatry 2021; 89:1116-1126. [PMID: 33722387 DOI: 10.1016/j.biopsych.2020.12.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/13/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022]
Abstract
Studying the stress response is a major pillar of neuroscience research not only because stress is a daily reality but also because the exquisitely fine-tuned bodily changes triggered by stress are a neuroendocrinological marvel. While the genome-wide changes induced by chronic stress have been extensively studied, we know surprisingly little about the complex molecular cascades triggered by acute stressors, the building blocks of chronic stress. The acute stress (or fight-or-flight) response mobilizes organismal energy resources to meet situational demands. However, successful stress coping also requires the efficient termination of the stress response. Maladaptive coping-particularly in response to severe or repeated stressors-can lead to allostatic (over)load, causing wear and tear on tissues, exhaustion, and disease. We propose that deep molecular profiling of the changes triggered by acute stressors could provide molecular correlates for allostatic load and predict healthy or maladaptive stress responses. We present a theoretical framework to interpret multiomic data in light of energy homeostasis and activity-dependent gene regulation, and we review the signaling cascades and molecular changes rapidly induced by acute stress in different cell types in the brain. In addition, we review and reanalyze recent data from multiomic screens conducted mainly in the rodent hippocampus and amygdala after acute psychophysical stressors. We identify challenges surrounding experimental design and data analysis, and we highlight promising new research directions to better understand the stress response on a multiomic level.
Collapse
Affiliation(s)
- Amalia Floriou-Servou
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Lukas von Ziegler
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Rebecca Waag
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Christa Schläppi
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland
| | - Pierre-Luc Germain
- Computational Neurogenomics, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland; Laboratory of Statistical Bioinformatics, Department for Molecular Life Sciences, University of Zürich, Zürich, Switzerland.
| | - Johannes Bohacek
- Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Switzerland; Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zürich, Switzerland.
| |
Collapse
|
5
|
Gadasheva Y, Nolze A, Grossmann C. Posttranslational Modifications of the Mineralocorticoid Receptor and Cardiovascular Aging. Front Mol Biosci 2021; 8:667990. [PMID: 34124152 PMCID: PMC8193679 DOI: 10.3389/fmolb.2021.667990] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/07/2021] [Indexed: 01/05/2023] Open
Abstract
During aging, the cardiovascular system is especially prone to a decline in function and to life-expectancy limiting diseases. Cardiovascular aging is associated with increased arterial stiffness and vasoconstriction as well as left ventricular hypertrophy and reduced diastolic function. Pathological changes include endothelial dysfunction, atherosclerosis, fibrosis, hypertrophy, inflammation, and changes in micromilieu with increased production of reactive oxygen and nitrogen species. The renin-angiotensin-aldosterone-system is an important mediator of electrolyte and blood pressure homeostasis and a key contributor to pathological remodeling processes of the cardiovascular system. Its effects are partially conveyed by the mineralocorticoid receptor (MR), a ligand-dependent transcription factor, whose activity increases during aging and cardiovascular diseases without correlating changes of its ligand aldosterone. There is growing evidence that the MR can be enzymatically and non-enzymatically modified and that these modifications contribute to ligand-independent modulation of MR activity. Modifications reported so far include phosphorylation, acetylation, ubiquitination, sumoylation and changes induced by nitrosative and oxidative stress. This review focuses on the different posttranslational modifications of the MR, their impact on MR function and degradation and the possible implications for cardiovascular aging and diseases.
Collapse
Affiliation(s)
- Yekatarina Gadasheva
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Nolze
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
6
|
Phasing the intranuclear organization of steroid hormone receptors. Biochem J 2021; 478:443-461. [DOI: 10.1042/bcj20200883] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/28/2020] [Accepted: 01/07/2021] [Indexed: 12/15/2022]
Abstract
Steroid receptors (SRs) encompass a family of transcription factors that regulate the expression of thousands of genes upon binding to steroid hormones and include the glucocorticoid, androgen, progesterone, estrogen and mineralocorticoid receptors. SRs control key physiological and pathological processes, thus becoming relevant drug targets. As with many other nuclear proteins, hormone-activated SRs concentrate in multiple discrete foci within the cell nucleus. Even though these foci were first observed ∼25 years ago, their exact structure and function remained elusive. In the last years, new imaging methodologies and theoretical frameworks improved our understanding of the intranuclear organization. These studies led to a new paradigm stating that many membraneless nuclear compartments, including transcription-related foci, form through a liquid–liquid phase separation process. These exciting ideas impacted the SR field by raising the hypothesis of SR foci as liquid condensates involved in transcriptional regulation. In this work, we review the current knowledge about SR foci formation under the light of the condensate model, analyzing how these structures may impact SR function. These new ideas, combined with state-of-the-art techniques, may shed light on the biophysical mechanisms governing the formation of SR foci and the biological function of these structures in normal physiology and disease.
Collapse
|
7
|
Effects of Maternal Chewing on Prenatal Stress-Induced Cognitive Impairments in the Offspring via Multiple Molecular Pathways. Int J Mol Sci 2020; 21:ijms21165627. [PMID: 32781547 PMCID: PMC7460630 DOI: 10.3390/ijms21165627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 12/14/2022] Open
Abstract
We aimed to investigate the effects of maternal chewing on prenatal stress-induced cognitive impairments in the offspring and to explore the molecular pathways of maternal chewing in a mice model. Maternal chewing ameliorated spatial learning impairments in the offspring in a Morris water maze test. Immunohistochemistry and Western blot findings revealed that maternal chewing alleviated hippocampal neurogenesis impairment and increased the expression of hippocampal brain-derived neurotrophic factor in the offspring. In addition, maternal chewing increased the expression of glucocorticoid receptor (GR) and 11β-hydroxysteroid dehydrogenase isozyme 2 (11β-HSD2) and decreased the expression of 11β-HSD1 in the placenta, thereby attenuating the increase of glucocorticoid in the offspring. Furthermore, maternal chewing increased the expression of 11β-HSD2, FK506-binding protein 51 (FKBP51) and FKBP52 and decreased the expression of 11β-HSD1, thereby increasing hippocampal nuclear GR level. In addition, maternal chewing attenuated the increase in expression of DNMT1 and DNMT3a and the decrease in expression of histone H3 methylation at lysine 4, 9, 27 and histone H3 acetylation at lysine 9 induced by prenatal stress in the offspring. Our findings suggest that maternal chewing could ameliorate prenatal stress-induced cognitive impairments in the offspring at least in part by protecting placenta barrier function, alleviating hippocampal nuclear GR transport impairment and increasing the hippocampal brain-derived neurotrophic factor (BDNF) level.
Collapse
|
8
|
Sotiropoulos I, Silva JM, Gomes P, Sousa N, Almeida OFX. Stress and the Etiopathogenesis of Alzheimer's Disease and Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:241-257. [PMID: 32096043 DOI: 10.1007/978-981-32-9358-8_20] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder with a complex physiopathology whose initiators are poorly defined. Accumulating clinical and experimental evidence suggests a causal role of lifetime stress in AD. This chapter summarizes current knowledge about how chronic stress and its accompanying high levels of glucocorticoid (GC) secretion, trigger the two main pathomechanisms of AD: (i) misprocessing of amyloid precursor protein (APP) and the generation of amyloid beta (Aβ) and (ii) Tau hyperphosphorylation and aggregation. Given that depression is a well-known stress-related illness, and the evidence that depression may precede AD, this chapter also explores neurobiological mechanisms that may be common to depressive and AD pathologies. This review also discusses emerging insights into the role of Tau and its malfunction in disrupting neuronal cascades and neuroplasticity and, thus triggering brain pathology.
Collapse
Affiliation(s)
- Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho - Campus de Gualtar, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Joana M Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho - Campus de Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patricia Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho - Campus de Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho - Campus de Gualtar, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | |
Collapse
|
9
|
Xiao X, Zhang H, Wang H, Li Q, Zhang T. Neuroprotective effect of amantadine on corticosterone-induced abnormal glutamatergic synaptic transmission of CA3-CA1 pathway in rat's hippocampal slices. Synapse 2017; 71. [PMID: 28902436 DOI: 10.1002/syn.22010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/04/2017] [Accepted: 09/08/2017] [Indexed: 01/11/2023]
Abstract
Depression is a psychiatric disorder and chronic stress, leading to altered glucocorticoid secretion patterns, is one of the factors that induce depression. Our previous study showed that amantadine significantly attenuated the impairments of synaptic plasticity and cognitive function a rat model of CUS. However, little is known regarding the underlying mechanism. In the present study, the whole-cell patch-clamp technique was applied to examine the protection effect of amantadine on the hippocampus CA3-CA1 pathway. Evoked excitatory postsynaptic currents (eEPSCs), miniature excitatory postsynaptic currents (mEPSCs), paired-pulse ratio (PPR) and the action potentials of CA3 neurons were recorded. Our data showed that corticosterone increased the amplitude of eEPSCs and decreased the value of paired-pulse ratio (PPR), but both of them were significantly reversed by amantadine. In addition, the frequency of mEPSC was considerably increased by corticosterone, but it was reduced by amantadine. Moreover, we used the Fluo-3/AM image to detect the Ca2+ influx in primary cultured hippocampal neurons. The results showed that the intracellular calcium levels were significantly decreased by amantadine in the corticosterone treated neurons. Additionally, the superoxide dismutase (SOD) and catalase (CAT) activities were reduced by corticosterone, while they were enhanced by either amantadine or low-calcium artificial cerebral spinal fluid (ACSF). These results suggest that amantadine significantly improves corticosterone-induced abnormal glutamatergic synaptic transmission of CA3-CA1 synapses presynaptically and alleviates the activities of antioxidant enzymes via regulating the calcium influx.
Collapse
Affiliation(s)
- Xi Xiao
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Hui Zhang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Hui Wang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Qun Li
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| | - Tao Zhang
- Department of Zoology and Developmental Biology, College of Life Sciences and Key Laboratory of Bioactive Materials Ministry of Education, Nankai University 300071, Tianjin, People's Republic of China
| |
Collapse
|
10
|
HuR-Dependent Editing of a New Mineralocorticoid Receptor Splice Variant Reveals an Osmoregulatory Loop for Sodium Homeostasis. Sci Rep 2017; 7:4835. [PMID: 28684740 PMCID: PMC5500589 DOI: 10.1038/s41598-017-04838-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/12/2017] [Indexed: 02/04/2023] Open
Abstract
Aldosterone and the Mineralocorticoid Receptor (MR) control hydroelectrolytic homeostasis and alterations of mineralocorticoid signaling pathway are involved in the pathogenesis of numerous human diseases, justifying the need to decipher molecular events controlling MR expression level. Here, we show in renal cells that the RNA-Binding Protein, Human antigen R (HuR), plays a central role in the editing of MR transcript as revealed by a RNA interference strategy. We identify a novel Δ6 MR splice variant, which lacks the entire exon 6, following a HuR-dependent exon skipping event. Using isoform-specific TaqMan probes, we show that Δ6 MR variant is expressed in all MR-expressing tissues and cells and demonstrate that extracelullar tonicity regulates its renal expression. More importantly, this splice variant exerts dominant-negative effects on transcriptional activity of the full-length MR protein. Collectively, our data highlight a crucial role of HuR as a master posttranscriptional regulator of MR expression in response to osmotic stress. We demonstrate that hypotonicity, not only enhances MR mRNA stability, but also decreases expression of the Δ6 MR variant, thus potentiating renal MR signaling. These findings provide compelling evidence for an autoregulatory feedback loop for the control of sodium homeostasis through posttranscriptional events, likely relevant in renal pathophysiological situations.
Collapse
|
11
|
Nishi M, Kawata M. Brain Corticosteroid Receptor Dynamics and Trafficking: Implications from Live Cell Imaging. Neuroscientist 2016; 12:119-33. [PMID: 16514009 DOI: 10.1177/1073858405279691] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Adrenal corticosteroids (cortisol in humans or corticosterone in rodents) exert numerous effects in the central nervous system that regulate the stress response, mood, learning and memory, and various neuroendocrine functions. Corticosterone actions in the brain are mediated by two corticosteroid receptors, glucocorticoid receptor (GR) and mineralocorticoid receptor (MR), and they show a high degree of colocalization in the hippocampal region. These receptors predominantly reside in the cytoplasm without ligand and are translocated into the nucleus upon ligand binding to act as transcriptional factors. Thus, their subcellualr localizations are an important component of their biological activity. Given the differential action of MR and GR in the central nervous system, it is important to elucidate how the trafficking of these receptors between the cytoplasm and the nucleus and their interactions are regulated by ligand or other molecules to exert transcriptional activity. In this review, the authors focus on the nucleocytoplasmic and subnuclear trafficking of GR and MR in neural cells and nonneural cells and discuss various factors affecting the dynamics of these receptors.
Collapse
Affiliation(s)
- Mayumi Nishi
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | | |
Collapse
|
12
|
Jiménez-Canino R, Lorenzo-Díaz F, Jaisser F, Farman N, Giraldez T, Alvarez de la Rosa D. Histone Deacetylase 6-Controlled Hsp90 Acetylation Significantly Alters Mineralocorticoid Receptor Subcellular Dynamics But Not its Transcriptional Activity. Endocrinology 2016; 157:2515-32. [PMID: 27100623 DOI: 10.1210/en.2015-2055] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mineralocorticoid receptor (MR) is a member of the nuclear receptor superfamily that transduces the biological effects of corticosteroids. Its best-characterized role is to enhance transepithelial sodium reabsorption in response to increased aldosterone levels. In addition, MR participates in other aldosterone- or glucocorticoid-controlled processes such as cardiovascular homeostasis, adipocyte differentiation or neurogenesis, and regulation of neuronal activity in the hippocampus. Like other steroid receptors, MR forms cytosolic heterocomplexes with heat shock protein (Hsp) 90), Hsp70, and other proteins such as immunophilins. Interaction with Hsp90 is thought to maintain MR in a ligand-binding competent conformation and to regulate ligand-dependent and -independent nucleocytoplasmatic shuttling. It has previously been shown that acetylation of residue K295 in Hsp90 regulates its interaction with the androgen receptor and glucocorticoid receptor (GR). In this work we hypothesized that Hsp90 acetylation provides a regulatory step to modulate MR cellular dynamics and activity. We used Hsp90 acetylation mimic mutant K295Q or nonacetylatable mutant K295R to examine whether MR nucleocytoplasmatic shuttling and gene transactivation are affected. Furthermore, we manipulated endogenous Hsp90 acetylation levels by controlling expression or activity of histone deacetylase 6 (HDAC6), the enzyme responsible for deacetylation of Hsp90-K295. Our data demonstrates that HDAC6-mediated Hsp90 acetylation regulates MR cellular dynamics but it does not alter its function. This stands in contrast with the down-regulation of GR by HDAC6, suggesting that Hsp90 acetylation may play a role in balancing relative MR and GR activity when both factors are co-expressed in the same cell.
Collapse
Affiliation(s)
- Rubén Jiménez-Canino
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| | - Fabián Lorenzo-Díaz
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| | - Frederic Jaisser
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| | - Nicolette Farman
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| | - Teresa Giraldez
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| | - Diego Alvarez de la Rosa
- Department of Physiology (R.J.-C., F.L.-D., T.G., D.A.d.l.R.), Institute of Biomedical Technologies and Center for Biomedical Research of the Canary Islands, University of La Laguna, Tenerife 38071, Spain; and INSERM UMRS 1138 (N.J., N.F.), Team 1, Centre de Recherche des Cordeliers, Université Pierre et Marie Curie, Paris 75006, France
| |
Collapse
|
13
|
Mani O, Nashev LG, Livelo C, Baker ME, Odermatt A. Role of Pro-637 and Gln-642 in human glucocorticoid receptors and Ser-843 and Leu-848 in mineralocorticoid receptors in their differential responses to cortisol and aldosterone. J Steroid Biochem Mol Biol 2016; 159:31-40. [PMID: 26907965 DOI: 10.1016/j.jsbmb.2016.02.017] [Citation(s) in RCA: 9] [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: 11/01/2015] [Revised: 02/11/2016] [Accepted: 02/18/2016] [Indexed: 10/22/2022]
Abstract
Mineralocorticoid receptors (MR) and glucocorticoid receptors (GR) are descended from a common ancestral corticoid receptor. The basis for specificities of human MR for aldosterone and human GR for glucocorticoids, such as cortisol, bearing 17α-hydroxyl-groups, is incompletely understood. Differences in MR at S843 and L848 and GR at the corresponding P637 and Q642 have been proposed as important in their different responses to glucocorticoids with 17α-hydroxyl-groups. We investigated the impact of these residues on binding affinity (Ki) and transcriptional activation (EC50) of mutants MR-S843P, MR-L848Q and MR-S843P/L848Q and mutants GR-P637S, GR-Q642L and GR-P637S/Q642L in the presence of different corticosteroids. Aldosterone, cortisol and corticosterone had similar affinities for wild-type MR and all mutants, while dexamethasone had increased affinity for the three mutants. However, transactivation of MR-S843P and MR-S843P/L848Q by all four steroids was significantly lower than for wild-type MR. In contrast, transactivation of MR-L848Q tended to be 3-fold higher for cortisol and corticosterone and increased 7-fold for dexamethasone, indicating that MR-L848Q has an increased response to glucocorticoids, while retaining a strong response to aldosterone. Compared to wild-type GR, GR-P637S and GR-Q642L had increased affinities and significantly increased transcriptional activity with aldosterone and corticosterone, and GR-P637S had similar transcriptional activity with cortisol and dexamethasone, while GR-Q642L and GR-P637S/Q642L had a significant decrease in transcriptional activity with cortisol and dexamethasone. 3D-models of these MR and GR mutants revealed that dexamethasone and aldosterone, respectively, fit nicely into the steroid-binding pocket, consistent with the affinity of dexamethasone for MR mutants and aldosterone for GR mutants.
Collapse
Affiliation(s)
- Orlando Mani
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Lyubomir G Nashev
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Christopher Livelo
- Department of Medicine, 0693, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Michael E Baker
- Department of Medicine, 0693, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States.
| | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, Pharmacenter, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland.
| |
Collapse
|
14
|
Abstract
The hypothalamo-pituitary-adrenal axis (HPA) is responsible for stimulation of adrenal corticosteroids in response to stress. Negative feedback control by corticosteroids limits pituitary secretion of corticotropin, ACTH, and hypothalamic secretion of corticotropin-releasing hormone, CRH, and vasopressin, AVP, resulting in regulation of both basal and stress-induced ACTH secretion. The negative feedback effect of corticosteroids occurs by action of corticosteroids at mineralocorticoid receptors (MR) and/or glucocorticoid receptors (GRs) located in multiple sites in the brain and in the pituitary. The mechanisms of negative feedback vary according to the receptor type and location within the brain-hypothalmo-pituitary axis. A very rapid nongenomic action has been demonstrated for GR action on CRH neurons in the hypothalamus, and somewhat slower nongenomic effects are observed in the pituitary or other brain sites mediated by GR and/or MR. Corticosteroids also have genomic actions, including repression of the pro-opiomelanocortin (POMC) gene in the pituitary and CRH and AVP genes in the hypothalamus. The rapid effect inhibits stimulated secretion, but requires a rapidly rising corticosteroid concentration. The more delayed inhibitory effect on stimulated secretion is dependent on the intensity of the stimulus and the magnitude of the corticosteroid feedback signal, but also the neuroanatomical pathways responsible for activating the HPA. The pathways for activation of some stressors may partially bypass hypothalamic feedback sites at the CRH neuron, whereas others may not involve forebrain sites; therefore, some physiological stressors may override or bypass negative feedback, and other psychological stressors may facilitate responses to subsequent stress.
Collapse
|
15
|
Le Menuet D, Lombès M. The neuronal mineralocorticoid receptor: from cell survival to neurogenesis. Steroids 2014; 91:11-9. [PMID: 24928721 DOI: 10.1016/j.steroids.2014.05.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 04/18/2014] [Accepted: 05/28/2014] [Indexed: 02/07/2023]
Abstract
Mineralocorticoid receptor (MR), a hormone-activated transcription factor belonging to the nuclear receptor superfamily, exerts widespread actions in many tissues such as tight epithelia, the cardiovascular system, adipose tissues and macrophages. In the mammalian brain, MR is present in the limbic areas where it is highly expressed in neurons of the hippocampus and mostly absent in other regions while the glucocorticoid receptor (GR) expression is ubiquitous. MR binds both aldosterone and glucocorticoids, the latter having a ten-fold higher affinity for MR than for the closely related GR. However, owing to the minimal aldosterone transfer across the blood brain barrier and the absence of neuronal 11β hydroxysteroid dehydrogenase type 2 as an intracellular gate-keeper, neuronal MR appears to be fully occupied even at low physiological glucocorticoid levels while GR activation only occurs at high glucocorticoid concentrations, i.e. at the peak of the circadian rhythm or under stress. This defined a one hormone/two receptors system that works in balance, modulating a large spectrum of actions in the central nervous system. MR and GR are involved in the stress responses, the regulation of neuron excitability, long term potentiation, neuroprotection and neurogenesis in the dentate gyrus. MR thus constitutes a key factor in the arising of higher cognitive functions such as memorization, learning and mood. This review presents an overview of various roles of MR in the central nervous system which are somewhat less studied than that of GR, in the light of recent data obtained using cellular models, animal models and clinical investigations.
Collapse
Affiliation(s)
- Damien Le Menuet
- Inserm U693, Le Kremlin-Bicêtre F-94276, France; Univ Paris-Sud, Faculté de Médecine Paris-Sud, UMR-S693, Le Kremlin-Bicêtre F-94276, France.
| | - Marc Lombès
- Inserm U693, Le Kremlin-Bicêtre F-94276, France; Univ Paris-Sud, Faculté de Médecine Paris-Sud, UMR-S693, Le Kremlin-Bicêtre F-94276, France; Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin Bicêtre F-94275, France
| |
Collapse
|
16
|
Abstract
The primary adrenal cortical steroid hormones, aldosterone, and the glucocorticoids cortisol and corticosterone, act through the structurally similar mineralocorticoid (MR) and glucocorticoid receptors (GRs). Aldosterone is crucial for fluid, electrolyte, and hemodynamic homeostasis and tissue repair; the significantly more abundant glucocorticoids are indispensable for energy homeostasis, appropriate responses to stress, and limiting inflammation. Steroid receptors initiate gene transcription for proteins that effect their actions as well as rapid non-genomic effects through classical cell signaling pathways. GR and MR are expressed in many tissues types, often in the same cells, where they interact at molecular and functional levels, at times in synergy, others in opposition. Thus the appropriate balance of MR and GR activation is crucial for homeostasis. MR has the same binding affinity for aldosterone, cortisol, and corticosterone. Glucocorticoids activate MR in most tissues at basal levels and GR at stress levels. Inactivation of cortisol and corticosterone by 11β-HSD2 allows aldosterone to activate MR within aldosterone target cells and limits activation of the GR. Under most conditions, 11β-HSD1 acts as a reductase and activates cortisol/corticosterone, amplifying circulating levels. 11β-HSD1 and MR antagonists mitigate inappropriate activation of MR under conditions of oxidative stress that contributes to the pathophysiology of the cardiometabolic syndrome; however, MR antagonists decrease normal MR/GR functional interactions, a particular concern for neurons mediating cognition, memory, and affect.
Collapse
Affiliation(s)
- Elise Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Celso E. Gomez-Sanchez
- G.V.(Sonny) Montgomery V.A. Medical Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
17
|
Han F, Ding J, Shi Y. Expression of amygdala mineralocorticoid receptor and glucocorticoid receptor in the single-prolonged stress rats. BMC Neurosci 2014; 15:77. [PMID: 24947040 PMCID: PMC4074391 DOI: 10.1186/1471-2202-15-77] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/09/2014] [Indexed: 01/10/2023] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is an anxious disorder associated with low levels of corticosterone and enhanced negative feedback of the hypothalamic–pituitary–adrenal (HPA) axis. Previous studies showed that the amygdala not only has an excitatory effect on the HPA axis but also plays a key role in fear-related behaviors. Coticosterone exert actions through binding to the mineralocorticoid (MR) and glucocorticoid receptor (GR), which are abundant in the amygdala. In our previous study, down-regulation of MR and GR in the hippocampus of PTSD rats was found. But the roles of MR and GR in the amygdala of PTSD rats is incompletely understood. Results wistar rats were divided into 1 d, 7 d, 14 d groups after single prolonged stress (SPS) and control group. SPS is a reliable animal model of PTSD. Open field test (OF) and elevated plus maze tests (EPM) were performed to examine fear-related behaviors. Morphological changes of the ultrastructure of the amygdala neurons were assessed by transmission electron microscopy (TEM). Dual-immunofluorescence histochemistry was used to determined subcellular distribution and colocalization of MR- and GR-ir. Protein and mRNA of MR and GR was examined by western blotting and RT-PCR. OF and EPM showed enhanced fear in SPS rats. Abnormal neuronal morphology was discovered in the amygdala of SPS rats. The expression of MR- and GR-ir intensity, mRNA and protein within the amygdala decreased after SPS at 1 day, and then gradually recovered by 14 days, although the degree of decrease and recovery were different amongst techniques. We found no change in the MR/GR ratio at 3 levels of the amygdala. But more cytoplasmic distribution and decreased colocalization of MR- and GR-ir were observed in the amygdala after 7 days of SPS. Conclusion These data suggest that change of MR and GR in the amygdala are involved in the mechanisms of fear in PTSD.
Collapse
Affiliation(s)
| | | | - Yuxiu Shi
- PTSD lab, Department of Histology and Embryology, Institute of pathology and Pathophysiology, China Medical University, Shenyang 110001, China.
| |
Collapse
|
18
|
Grossmann C, Ruhs S, Langenbruch L, Mildenberger S, Strätz N, Schumann K, Gekle M. Nuclear shuttling precedes dimerization in mineralocorticoid receptor signaling. ACTA ACUST UNITED AC 2014; 19:742-51. [PMID: 22726688 DOI: 10.1016/j.chembiol.2012.04.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 04/22/2012] [Accepted: 04/25/2012] [Indexed: 10/28/2022]
Abstract
The mineralocorticoid receptor (MR), a member of the steroid receptor superfamily, regulates water-electrolyte balance and mediates pathophysiological effects in the renocardiovascular system. Previously, it was assumed that after binding aldosterone, the MR dissociates from HSP90, forms homodimers, and then translocates into the nucleus where it acts as a transcription factor (Guiochon-Mantel et al., 1989; Robertson et al., 1993; Savory et al., 2001). We found that, during aldosterone-induced nuclear translocation, MR is bound to HSP90 both in the cytosol and the nucleus. Homodimerization measured by eBRET and FRET takes place when the MR is already predominantly nuclear. In vitro binding of MR to DNA was independent of ligand but could be partially inhibited by geldanamycin. Overall, here we provide insights into classical MR signaling necessary for elucidating the mechanisms of pathophysiological MR effects and MR specificity.
Collapse
Affiliation(s)
- Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, University Halle-Wittenberg, Halle/Saale 06112, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
The mineralocorticoid receptor (MR) is a ligand-dependent transcription factor that physiologically regulates water-electrolyte homeostasis and controls blood pressure. The MR can also elicit inflammatory and remodeling processes in the cardiovascular system and the kidneys, which require the presence of additional pathological factors like for example nitrosative stress. However, the underlying molecular mechanism(s) for pathophysiological MR effects remain(s) elusive. The inactive MR is located in the cytosol associated with chaperone molecules including HSP90. After ligand binding, the MR monomer rapidly translocates into the nucleus while still being associated to HSP90 and after dissociation from HSP90 binds to hormone-response-elements called glucocorticoid response elements (GREs) as a dimer. There are indications that rapid MR trafficking is modulated in the presence of high salt, oxidative or nitrosative stress, hypothetically by induction or posttranslational modifications. Additionally, glucocorticoids and the enzyme 11beta hydroxysteroid dehydrogenase may also influence MR activation. Because MR trafficking and its modulation by micro-milieu factors influence MR cellular localization, it is not only relevant for genomic but also for nongenomic MR effects.
Collapse
Affiliation(s)
- M Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - M Bretschneider
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - S Meinel
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - S Ruhs
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany
| | - C Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Germany.
| |
Collapse
|
20
|
Caudal D, Jay TM, Godsil BP. Behavioral stress induces regionally-distinct shifts of brain mineralocorticoid and glucocorticoid receptor levels. Front Behav Neurosci 2014; 8:19. [PMID: 24523684 PMCID: PMC3905199 DOI: 10.3389/fnbeh.2014.00019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 01/13/2014] [Indexed: 12/23/2022] Open
Abstract
Mineralocorticoid and glucocorticoid receptors (MRs and GRs) mediate the impact of stress on brain function primarily by affecting gene transcription in the cell nucleus. In vitro studies using hippocampal neurons indicate that MRs and GRs translocate to the nucleus after binding to the stress hormone corticosterone, yet the in vivo temporal dynamics of MR and GR levels in other limbic regions critical for the stress response, however, are largely unknown. Rats underwent an elevated platform (EP) stress procedure and brain tissue was sampled from the amygdala (AMY), medial prefrontal cortex (mPFC), dorsal hippocampus and ventral hippocampus. By measuring MR and GR levels in the nuclear fraction from the tissue sampled, we observed striking shifts in the protein levels that varied by receptor, brain region and by the time after EP stress. These findings indicate that the subcellular trafficking of corticosteroid receptors display distinct temporal dynamics in different limbic regions after behavioral stress. These heterogeneous effects could underlie contrasting regional responses to stress within the brain, and they highlight the importance for systems-level analysis of stress responsivity.
Collapse
Affiliation(s)
- Dorian Caudal
- Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences U894, INSERM Paris, France ; Faculté de Médecine Paris Descartes, Université Paris Descartes Paris, France
| | - Thérèse M Jay
- Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences U894, INSERM Paris, France ; Faculté de Médecine Paris Descartes, Université Paris Descartes Paris, France
| | - Bill P Godsil
- Laboratoire de Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences U894, INSERM Paris, France ; Faculté de Médecine Paris Descartes, Université Paris Descartes Paris, France
| |
Collapse
|
21
|
Caldwell KK, Goggin SL, Tyler CR, Allan AM. Prenatal alcohol exposure is associated with altered subcellular distribution of glucocorticoid and mineralocorticoid receptors in the adolescent mouse hippocampal formation. Alcohol Clin Exp Res 2013; 38:392-400. [PMID: 23992407 PMCID: PMC3864567 DOI: 10.1111/acer.12236] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 07/06/2013] [Indexed: 11/29/2022]
Abstract
Background Accumulating evidence indicates that several of the long-term consequences of prenatal alcohol exposure (PAE) are the result of changes in the development and function of cortico-limbic structures, including the hippocampal formation. The glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) are key regulators of hippocampal formation development, structure, and functioning and, thus, are potential mediators of PAE’s effects on this brain region. In the present studies, we assessed the impact of PAE on components of corticosteroid signaling pathways in the mouse hippocampal formation. Methods Throughout pregnancy, mouse dams were offered either 10% (w/v) ethanol sweetened with 0.066% (w/v) saccharin (SAC) or 0.066% (w/v) SAC alone using a limited (4-hour) access, drinking-in-the-dark paradigm. The hippocampal formation was isolated from naïve postnatal day 40 to 50 offspring, and subcellular fractions were prepared. Using immunoblotting techniques, we measured the levels of GR, MR, 11-β-hydroxysteroid dehydrogenase 1 (11β-HSD1), and the FK506-binding proteins 51 (FKBP51, FKBP5) and 52 (FKBP52, FKBP4). Finally, we determined the effect of PAE on context discrimination, a hippocampal-dependent learning/memory task. Results PAE was associated with reduced MR and elevated GR nuclear localization in the hippocampal formation, whereas cytosolic levels of both receptors were not significantly altered. FKBP51 levels were reduced, while FKBP52 levels were unaltered, and 11β-HSD1 levels were increased in postnuclear fractions isolated from PAE mouse hippocampal formation. These neurochemical alterations were associated with reduced context discrimination. Conclusions The data support a model in which PAE leads to increased nuclear localization of GRs secondary to reductions in FKBP51 and increases in 11β-HSD1 levels in the adolescent mouse hippocampal formation. Persistent dysregulation of GR subcellular distribution is predicted to damage the hippocampal formation and may underlie many of the effects of PAE on hippocampal-dependent functioning.
Collapse
Affiliation(s)
- Kevin K Caldwell
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | | | | | | |
Collapse
|
22
|
Robertson S, Hapgood JP, Louw A. Glucocorticoid receptor concentration and the ability to dimerize influence nuclear translocation and distribution. Steroids 2013. [PMID: 23178279 DOI: 10.1016/j.steroids.2012.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Glucocorticoid receptor (GR) concentrations and the ability of the GR to dimerize are factors which influence sensitivity to glucocorticoids. Upon glucocorticoid binding, the GR is actively transported into the nucleus, a crucial step in determining GR function. We examined the effects of GR concentration and the ability to dimerize on GR nuclear import, export and nuclear distribution using both live cell microscopy of GFP-tagged GR and immunofluorescence of untagged GR, with both wild type GR (GRwt) and dimerization deficient GR (GRdim). We found that the observed rate of GR nuclear import increases significantly at higher GR concentrations, at saturating concentrations of dexamethasone (10(-6) M) using GFP-tagged GR, while with untagged GR it is only discernable at sub-saturating ligand concentrations (10(-10)-10(-9) M). Loss of dimerization results in a slower observed rate of nuclear import (2.5- to 3.3-fold decrease for GFP-GRdim) as well as a decreased extent of GR nuclear localization (18-27% decrease for untagged GRdim). These results were linked to an increased rate of GR export at low GR concentrations (1.4- to 1.6-fold increase for untagged GR) and where GR dimerization is abrogated (1.5- to 1.7-fold increase for GFP-GRdim). Furthermore, GR dimerization was shown to be required for the appearance of discrete GC-dependent GR nuclear foci, the loss of which may explain the increased rate of GR export for the GRdim. The reduction in the observed rate of nuclear import and increased rate of nuclear export displayed at low GR concentrations and by the GRdim could explain the lowered glucocorticoid response under these conditions.
Collapse
Affiliation(s)
- Steven Robertson
- Department of Biochemistry, University of Stellenbosch, Stellenbosch 7602, South Africa
| | | | | |
Collapse
|
23
|
Aguilar-Sánchez C, Hernández-Díaz I, Lorenzo-Díaz F, Navarro JF, Hughes TE, Giraldez T, Alvarez de la Rosa D. Identification of permissive insertion sites for generating functional fluorescent mineralocorticoid receptors. Endocrinology 2012; 153:3517-25. [PMID: 22621960 DOI: 10.1210/en.2012-1210] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The mineralocorticoid receptor (MR), a member of the nuclear receptor superfamily of transcription factors, is activated by aldosterone and mediates its natriferic action in tight epithelia. MR is also expressed in nonepithelial tissues. Importantly, it mediates the deleterious effects of inappropriately high aldosterone levels in the heart, in which it induces the development of cardiac fibrosis. Antagonism of MR in humans is useful in the treatment of severe cardiac failure and some forms of hypertension. Despite the important pathophysiological and pharmacological role of this receptor, many important questions about its cellular biology and functional roles remain unanswered. A major challenge in the study of MR is the unavailability of fully functional fluorescent derivatives of the receptor. In this study we have created a library of MR mutants with insertions of the yellow fluorescent protein in various internal locations in the receptor using a random-insertion transposon-based technique. Screening of this library using a transactivation assay allowed us to identify several fluorescent constructs that retain functionality. Detailed characterization of one of these construct showed that it induces aldosterone-target genes such as the epithelial Na(+) channel subunits and the serum and glucocorticoid-induced kinase 1 at physiological concentrations of aldosterone to an equal extent than the wild-type receptor. Furthermore, aldosterone affinity, hormone-induced nuclear translocation, DNA binding and regulation of nongenomic pathways are all indistinguishable from the wild-type receptor. This new set of fluorescent MR derivatives provides a useful tool for studying the cell biology of the receptor.
Collapse
Affiliation(s)
- Cristina Aguilar-Sánchez
- Department of Physiology and Instituto de Tecnologías Biomédicas, University of La Laguna, 38071 Tenerife, Spain
| | | | | | | | | | | | | |
Collapse
|
24
|
Wu H, Chen L, Zhou Q, Zhang W. AF17 facilitates Dot1a nuclear export and upregulates ENaC-mediated Na+ transport in renal collecting duct cells. PLoS One 2011; 6:e27429. [PMID: 22087315 PMCID: PMC3210795 DOI: 10.1371/journal.pone.0027429] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/16/2011] [Indexed: 01/22/2023] Open
Abstract
Our previous work in 293T cells and AF17-/- mice suggests that AF17 upregulates expression and activity of the epithelial Na+ channel (ENaC), possibly by relieving Dot1a-AF9-mediated repression. However, whether and how AF17 directly regulates Dot1a cellular distribution and ENaC function in renal collecting duct cells remain unaddressed. Here, we report our findings in mouse cortical collecting duct M-1 cells that overexpression of AF17 led to preferential distribution of Dot1a in the cytoplasm. This effect could be blocked by nuclear export inhibitor leptomycin B. siRNA-mediated depletion of AF17 caused nuclear accumulation of Dot1a. AF17 overexpression elicited multiple effects that are reminiscent of aldosterone action. These effects include 1) increased mRNA and protein expression of the three ENaC subunits (α, β and γ) and serum- and glucocorticoid inducible kinase 1, as revealed by real-time RT-qPCR and immunoblotting analyses; 2) impaired Dot1a-AF9 interaction and H3 K79 methylation at the αENaC promoter without affecting AF9 binding to the promoter, as evidenced by chromatin immunoprecipitation; and 3) elevated ENaC-mediated Na+ transport, as analyzed by measurement of benzamil-sensitive intracellular [Na+] and equivalent short circuit current using single-cell fluorescence imaging and an epithelial Volt-ohmmeter, respectively. Knockdown of AF17 elicited opposite effects. However, combination of AF17 overexpression or depletion with aldosterone treatment did not cause an additive effect on mRNA expression of the ENaC subunits. Taken together, we conclude that AF17 promotes Dot1a nuclear export and upregulates basal, but not aldosterone-stimulated ENaC expression, leading to an increase in ENaC-mediated Na+ transport in renal collecting duct cells.
Collapse
Affiliation(s)
- Hongyu Wu
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Lihe Chen
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Qiaoling Zhou
- Department of Internal Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Wenzheng Zhang
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Nishi M. Dynamics of corticosteroid receptors: lessons from live cell imaging. Acta Histochem Cytochem 2011; 44:1-7. [PMID: 21448312 PMCID: PMC3061448 DOI: 10.1267/ahc.10028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 10/05/2010] [Indexed: 01/24/2023] Open
Abstract
Adrenal corticosteroids (cortisol in humans or corticosterone in rodents) exert numerous effects on the central nervous system that regulates the stress response, mood, learning and memory, and various neuroendocrine functions. Corticosterone (CORT) actions in the brain are mediated via two receptor systems: the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). It has been shown that GR and MR are highly colocalized in the hippocampus. These receptors are mainly distributed in the cytoplasm without hormones and translocated into the nucleus after treatment with hormones to act as transcriptional factors. Thus the subcellular dynamics of both receptors are one of the most important issues. Given the differential action of MR and GR in the central nervous system, it is of great consequence to clarify how these receptors are trafficked between cytoplasm and nucleus and their interactions are regulated by hormones and/or other molecules to exert their transcriptional activity. In this review, we focus on the nucleocytoplasmic and subnuclear trafficking of GR and MR in neural cells and non-neural cells analyzed by using molecular imaging techniques with green fluorescent protein (GFP) including fluorescence recovery after photobleaching (FRAP) and fluorescence resonance energy transfer (FRET), and discuss various factors affecting the dynamics of these receptors. Furthermore, we discuss the future directions of in vivo molecular imaging of corticosteroid receptors at the whole brain level.
Collapse
Affiliation(s)
- Mayumi Nishi
- Department of Anatomy and Cell Biology, Nara Medical University
| |
Collapse
|
26
|
Ackermann D, Gresko N, Carrel M, Loffing-Cueni D, Habermehl D, Gomez-Sanchez C, Rossier BC, Loffing J. In vivo nuclear translocation of mineralocorticoid and glucocorticoid receptors in rat kidney: differential effect of corticosteroids along the distal tubule. Am J Physiol Renal Physiol 2010; 299:F1473-85. [DOI: 10.1152/ajprenal.00437.2010] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aldosterone and corticosterone bind to mineralocorticoid (MR) and glucocorticoid receptors (GR), which, upon ligand binding, are thought to translocate to the cell nucleus to act as transcription factors. Mineralocorticoid selectivity is achieved by the 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD2) that inactivates 11β-hydroxy glucocorticoids. High expression levels of 11β-HSD2 characterize the aldosterone-sensitive distal nephron (ASDN), which comprises the segment-specific cells of late distal convoluted tubule (DCT2), connecting tubule (CNT), and collecting duct (CD). We used MR- and GR-specific antibodies to study localization and regulation of MR and GR in kidneys of rats with altered plasma aldosterone and corticosterone levels. In control rats, MR and GR were found in cell nuclei of thick ascending limb (TAL), DCT, CNT, CD cells, and intercalated cells (IC). GR was also abundant in cell nuclei and the subapical compartment of proximal tubule (PT) cells. Dietary NaCl loading, which lowers plasma aldosterone, caused a selective removal of GR from cell nuclei of 11β-HSD2-positive ASDN. The nuclear localization of MR was unaffected. Adrenalectomy (ADX) resulted in removal of MR and GR from the cell nuclei of all epithelial cells. Aldosterone replacement rapidly relocated the receptors in the cell nuclei. In ASDN cells, low-dose corticosterone replacement caused nuclear localization of MR, but not of GR. The GR was redistributed to the nucleus only in PT, TAL, early DCT, and IC that express no or very little 11β-HSD2. In ASDN cells, nuclear GR localization was only achieved when corticosterone was replaced at high doses. Thus ligand-induced nuclear translocation of MR and GR are part of MR and GR regulation in the kidney and show remarkable segment- and cell type-specific characteristics. Differential regulation of MR and GR may alter the level of heterodimerization of the receptors and hence may contribute to the complexity of corticosteroid effects on ASDN function.
Collapse
Affiliation(s)
- Daniel Ackermann
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg
- Clinic for Nephrology and Hypertension, University Hospital of Bern, Bern
| | | | | | | | | | - Celso Gomez-Sanchez
- Department for Endocrinology, G. V. (Sonny) Montgomery Veterans Affairs Medical Center, University of Mississippi, Jackson, Mississippi; and
| | - Bernard C. Rossier
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Johannes Loffing
- Unit of Anatomy, Department of Medicine, University of Fribourg, Fribourg
- Institute for Anatomy, University of Zürich, Zürich
| |
Collapse
|
27
|
Hernández-Díaz I, Giraldez T, Arnau MR, Smits VAJ, Jaisser F, Farman N, Alvarez de la Rosa D. The mineralocorticoid receptor is a constitutive nuclear factor in cardiomyocytes due to hyperactive nuclear localization signals. Endocrinology 2010; 151:3888-99. [PMID: 20484457 DOI: 10.1210/en.2010-0099] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mineralocorticoid receptor (MR), a member of the nuclear receptor family, mediates the action of aldosterone in target epithelia, enhancing sodium reabsorption. In addition, MR may have other physiological functions in nonepithelial tissues. Altered expression or inappropriate activation of cardiac MR is directly linked to the development of cardiac fibrosis, and MR blockade is beneficial for the treatment of heart failure. However, the physiological role, activation status, and target genes of MR in the heart are poorly known. Because ligand-free steroid receptors are typically cytoplasmic and translocate to the nucleus upon ligand binding, we examined the subcellular localization of MR under different corticosteroid levels using subcellular fractionation and immunostaining. Our results demonstrate that MR is a chromatin-bound factor in mouse left ventricle and in a cultured model of cardiomyocytes, HL-1 cells, regardless of circulating corticosteroid levels. Immunohistochemical localization of MR in human heart confirms the subcellular localization pattern. Mutation of nuclear localization signals (NLSs) demonstrates that MR constitutive nuclear localization mainly depends on the synergistic contribution of NLS0 and NLS1. Constitutive nuclear localization in HL-1 cells can be reverted by cotransfection of heat shock protein 90. Heat shock protein 90 expression levels in the mouse heart and HL-1 cells are lower than those found in other tissues, suggesting that low levels of cochaperones render MR NLSs hyperactive in cardiomyocytes. Even though MR is constitutively nuclear, corticosteroids still control the transactivation properties of the receptor in a model promoter, although other MR ligand-independent activities cannot be excluded.
Collapse
Affiliation(s)
- Iván Hernández-Díaz
- Departamento de Fisiología, Facultad de Medicina, Universidad de La Laguna, La Laguna 38071, Spain
| | | | | | | | | | | | | |
Collapse
|
28
|
Reisenauer MR, Wang SW, Xia Y, Zhang W. Dot1a contains three nuclear localization signals and regulates the epithelial Na+ channel (ENaC) at multiple levels. Am J Physiol Renal Physiol 2010; 299:F63-76. [PMID: 20427473 PMCID: PMC2904171 DOI: 10.1152/ajprenal.00105.2010] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 04/21/2010] [Indexed: 12/14/2022] Open
Abstract
We have previously reported that Dot1a is located in the cytoplasm and nucleus (Reisenauer MR, Anderson M, Huang L, Zhang Z, Zhou Q, Kone BC, Morris AP, Lesage GD, Dryer SE, Zhang W. J Biol Chem 284: 35659-35669, 2009), widely expressed in the kidney as detected by its histone H3K79 methyltransferase activity (Zhang W, Hayashizaki Y, Kone BC. Biochem J 377: 641-651, 2004), and involved in transcriptional control of the epithelial Na(+) channel subunit-alpha gene (alphaENaC) (Zhang W, Xia X, Jalal DI, Kuncewicz T, Xu W, Lesage GD, Kone BC. Am J Physiol Cell Physiol 290: C936-C946, 2006). Aldosterone releases repression of alphaENaC by reducing expression of Dot1a and its partner AF9 (Zhang W, Xia X, Reisenauer MR, Hemenway CS, Kone BC. J Biol Chem 281: 18059-18068, 2006) and by impairing Dot1a-AF9 interaction via Sgk1-mediated AF9 phosphorylation (Zhang W, Xia X, Reisenauer MR, Rieg T, Lang F, Kuhl D, Vallon V, Kone BC. J Clin Invest 117: 773-783, 2007). This network also appears to regulate transcription of several other aldosterone target genes. Here, we provide evidence showing that Dot1a contains at least three potential nuclear localization signals (NLSs). Deletion of these NLSs causes green fluorescent protein-fused Dot1a fusions to localize almost exclusively in the cytoplasm of 293T cells as revealed by confocal microscopy. Deletion of NLSs abolished Dot1a-mediated repression of alphaENaC-promoter luciferase construct in M1 cells. AF9 is widely expressed in mouse kidney. Similar to alphaENaC, the mRNA levels of betaENaC, gammaENaC, and Sgk1 are also downregulated by Dot1a and AF9 overexpression. Small interference RNA-mediated knockdown of Dot1a and AF9 or aldosterone treatment leads to an opposite effect. Using single-cell fluorescence imaging or equivalent short-circuit current in IMCD3 and M1 cells, we show that observed transcriptional alterations correspond to changes in ENaC and Sgk1 protein levels as well as benzamil-sensitive Na(+) transport. In brief, Dot1a and AF9 downregulate Na(+) transport, most likely by regulating ENaC mRNA and subsequent protein expression and ENaC activity.
Collapse
Affiliation(s)
- Mary Rose Reisenauer
- Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
29
|
Cadwallader AB, Rollins DE, Lim CS. Effect of anabolic-androgenic steroids and glucocorticoids on the kinetics of hAR and hGR nucleocytoplasmic translocation. Mol Pharm 2010; 7:689-98. [PMID: 20230007 PMCID: PMC2882525 DOI: 10.1021/mp900259w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Although the qualitative nucleocytoplasmic transport of nuclear hormone receptors (NHRs) has been studied, there is little documentation of the cellular kinetics of this transport. Here, translocation studies using the human androgen receptor (hAR) and the human glucocorticoid receptor (hGR) were performed to aid in identifying the mechanism by which anabolic-androgenic steroids (AAS) were activating hAR and potentially interacting with hGR and how glucocorticoid ligands were interacting with the hGR and hAR. The real-time analysis of EGFP-labeled hAR and hGR ligand-induced cytoplasm-to-nucleus translocation was performed using fluorescence microscopy to better understand the action of these NHRs in a physiologically relevant cell-based model. After transient transfection, the hAR and hGR individually translocate as expected (i.e., transport is ligand-induced and dose-dependent) in this model biological system. Testosterone (TEST) had the fastest translocation rate for the hAR of 0.0525 min(-1). The other endogenous steroids, androstenedione (ANE) and dihydrotestosterone (DHT), had considerably lower hAR transport rates. The rates of hAR transport for the exogenous steroids methyltrienelone (MET), nandrolone (NAN), and oxandrolone (OXA) are lower than that of testosterone and similar to those of the endogenous steroids ANE and DHT. The hGR transport rates for cortisol (COR) and dexamethasone (DEX) are also presented. The synthetic GC, DEX, had a more rapid translocation rate (0.1599 min(-1)) at the highest dose of 100 nM compared to the endogenous GC COR (0.0431 min(-1)). The data obtained agrees with the existing qualitative data and adds an important ligand-dependent kinetic component to hAR and hGR transport. These kinetic data can aid our understanding of NHR action and interaction with other regulatory proteins, and can be useful in the development of new therapies.
Collapse
Affiliation(s)
- Amy B. Cadwallader
- Center for Human Toxicology, University of Utah Department of Pharmacology and Toxicology, 417 Wakara Way Suite 2111, Salt Lake City, UT, U.S.A. 84108
| | - Douglas E. Rollins
- Center for Human Toxicology, University of Utah Department of Pharmacology and Toxicology, 417 Wakara Way Suite 2111, Salt Lake City, UT, U.S.A. 84108
| | - Carol S. Lim
- University of Utah Department of Pharmaceutics and Pharmaceutical Chemistry, 421 Wakara Way Room 318, Salt Lake City, UT, U.S.A. 84108
| |
Collapse
|
30
|
Munier M, Meduri G, Viengchareun S, Leclerc P, Le Menuet D, Lombès M. Regulation of mineralocorticoid receptor expression during neuronal differentiation of murine embryonic stem cells. Endocrinology 2010; 151:2244-54. [PMID: 20207834 PMCID: PMC3107824 DOI: 10.1210/en.2009-0753] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Mineralocorticoid receptor (MR) plays a critical role in brain function. However, the regulatory mechanisms controlling neuronal MR expression that constitutes a key element of the hormonal response are currently unknown. Two alternative P1 and P2 promoters drive human MR gene transcription. To examine promoter activities and their regulation during neuronal differentiation and in mature neurons, we generated stably transfected recombinant murine embryonic stem cell (ES) lines, namely P1-GFP and P2-GFP, in which each promoter drove the expression of the reporter gene green fluorescent protein (GFP). An optimized protocol, using embryoid bodies and retinoic acid, permitted us to obtain a reproducible neuronal differentiation as revealed by the decrease in phosphatase alkaline activity, the concomitant appearance of morphological changes (neurites), and the increase in the expression of neuronal markers (nestin, beta-tubulin III, and microtubule-associated protein-2) as demonstrated by immunocytochemistry and quantitative PCR. Using these cell-based models, we showed that MR expression increased by 5-fold during neuronal differentiation, MR being preferentially if not exclusively expressed in mature neurons. Although the P2 promoter was always weaker than the P1 promoter during neuronal differentiation, their activities increased by 7- and 5-fold, respectively, and correlated with MR expression. Finally, although progesterone and dexamethasone were ineffective, aldosterone stimulated both P1 and P2 activity and MR expression, an effect that was abrogated by knockdown of MR by small interfering RNA. In conclusion, we provide evidence for a tight transcriptional control of MR expression during neuronal differentiation. Given the neuroprotective and antiapoptotic role proposed for MR, the neuronal differentiation of ES cell lines opens potential therapeutic perspectives in neurological and psychiatric diseases.
Collapse
Affiliation(s)
- Mathilde Munier
- Récepteurs stéroïdiens : physiopathologie endocrinienne et métabolique
INSERM : U693IFR93Université Paris Sud - Paris XIFaculté de médecine 63, Rue Gabriel Peri 94276 Le Kremlin Bicêtre,FR
| | - Geri Meduri
- Récepteurs stéroïdiens : physiopathologie endocrinienne et métabolique
INSERM : U693IFR93Université Paris Sud - Paris XIFaculté de médecine 63, Rue Gabriel Peri 94276 Le Kremlin Bicêtre,FR
- Service de génétique moléculaire, pharmacogénétique et hormonologie
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital BicêtreUniversité Paris Sud - Paris XI78, rue du Général Leclerc 94275 Le Kremlin Bicêtre,FR
| | - Say Viengchareun
- Récepteurs stéroïdiens : physiopathologie endocrinienne et métabolique
INSERM : U693IFR93Université Paris Sud - Paris XIFaculté de médecine 63, Rue Gabriel Peri 94276 Le Kremlin Bicêtre,FR
| | - Phillipe Leclerc
- IFR de Bicêtre
INSERM : IFR93Assistance publique - Hôpitaux de Paris (AP-HP)Université Paris Sud - Paris XIBatiment Inserm Gregory Pincus PARIS XI 80, Rue du General Leclerc 94276 Le Kremlin Bicêtre CEDEX,FR
| | - Damien Le Menuet
- Récepteurs stéroïdiens : physiopathologie endocrinienne et métabolique
INSERM : U693IFR93Université Paris Sud - Paris XIFaculté de médecine 63, Rue Gabriel Peri 94276 Le Kremlin Bicêtre,FR
| | - Marc Lombès
- Récepteurs stéroïdiens : physiopathologie endocrinienne et métabolique
INSERM : U693IFR93Université Paris Sud - Paris XIFaculté de médecine 63, Rue Gabriel Peri 94276 Le Kremlin Bicêtre,FR
- Service d'Endocrinologie et Maladies de la reproduction
Assistance publique - Hôpitaux de Paris (AP-HP)Hôpital BicêtreLe Kremlin Bicêtre 94275,FR
- * Correspondence should be adressed to: Marc Lombès
| |
Collapse
|
31
|
Proszkowiec-Weglarz M, Porter TE. Functional characterization of chicken glucocorticoid and mineralocorticoid receptors. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1257-68. [DOI: 10.1152/ajpregu.00805.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Glucocorticoid (GR) and mineralocorticoid (MR) receptors are ligand-activated transcription factors that belong to the nuclear hormone receptor superfamily. Little is known about the function of GR and MR in avian species. Recently, the chicken homologue of the GR (cGR) gene was cloned, and its tissue-specific expression was characterized, whereas the full-length sequence of the chicken MR (cMR) gene remains unknown. Therefore, the aims of this project were to clone the full-length cMR and to functionally characterize both chicken receptors. Cos-7 cells were transiently transfected with cGR or cMR expression vectors along with a glucocorticoid response element-luciferase (GRE-Luc) reporter construct. Transfected cells were then treated with increasing doses of corticosterone (CORT) or aldosterone (ALDO) alone and with GR or MR antagonists (ZK98299 and spironolactone, respectively). Transactivation of cGR or cMR was evaluated by luciferase assay. CORT and ALDO induced cGR- and cMR-driven transcriptional activity in a dose-dependent manner. Each receptor responded to both steroids, but cMR transcriptional activity was induced by lower levels of CORT and ALDO than cGR. Coexpression of both chicken corticosteroid receptors in Cos-7 cells had no synergistic or additive effect on CORT- or ALDO-induced transcriptional activity. Corticosteroid-dependent transactivation of cGR and cMR was partially blocked by antagonists. ZK98299 showed high specificity to cGR, while spironolactone had agonist properties toward both receptors. Immunocytochemistry was used to assess the cellular localization of both receptors. Corticosteroids induced translocation of both receptors into the nucleus. The functional properties of cGR and cMR determined in this study will be helpful in defining the physiological roles of GR and MR in avian species.
Collapse
Affiliation(s)
| | - Tom E. Porter
- University of Maryland, Department of Animal and Avian Sciences, College Park, Maryland
| |
Collapse
|
32
|
The hsp90-FKBP52 complex links the mineralocorticoid receptor to motor proteins and persists bound to the receptor in early nuclear events. Mol Cell Biol 2009; 30:1285-98. [PMID: 20038533 DOI: 10.1128/mcb.01190-09] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we demonstrate that the subcellular localization of the mineralocorticoid receptor (MR) is regulated by tetratricopeptide domain (TPR) proteins. The high-molecular-weight immunophilin (IMM) FKBP52 links the MR-hsp90 complex to dynein/dynactin motors favoring the cytoplasmic transport of MR to the nucleus. Replacement of this hsp90-binding IMM by FKBP51 or the TPR peptide favored the cytoplasmic localization of MR. The complete movement machinery, including dynein and tubulin, could be recovered from paclitaxel/GTP-stabilized cytosol and was fully reassembled on stripped MR immune pellets. The whole MR-hsp90-based heterocomplex was transiently recovered in the soluble fraction of the nucleus after 10 min of incubation with aldosterone. Moreover, cross-linked MR-hsp90 heterocomplexes accumulated in the nucleus in a hormone-dependent manner, demonstrating that the heterocomplex can pass undissociated through the nuclear pore. On the other hand, a peptide that comprises the DNA-binding domain of MR impaired the nuclear export of MR, suggesting the involvement of this domain in the process. This study represents the first report describing the entire molecular system that commands MR nucleocytoplasmic trafficking and proposes that the MR-hsp90-TPR protein heterocomplex is dissociated in the nucleus rather than in the cytoplasm.
Collapse
|
33
|
Alfarez DN, De Simoni A, Velzing EH, Bracey E, Joëls M, Edwards FA, Krugers HJ. Corticosterone reduces dendritic complexity in developing hippocampal CA1 neurons. Hippocampus 2009; 19:828-36. [PMID: 19235231 DOI: 10.1002/hipo.20566] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Although prolonged stress and corticosteroid exposure induce morphological changes in the hippocampal CA3 area, the adult CA1 area is quite resistant to such changes. Here we addressed the question whether elevated corticosteroid hormone levels change dendritic complexity in young, developing CA1 cells. In organotypic cultures (prepared from P5 rats) that were 14-21 days cultured in vitro, two doses of corticosterone (30 and 100 nM) were tested. Dendritic morphology of CA1 neurons was established by imaging neurons filled with the fluorescent dye Alexa. Application of 100 nM corticosterone for 20 minutes induced atrophy of the apical dendritic tree 1-4 hours later. Fractal analysis showed that total neuronal complexity was reduced twofold when compared with vehicle-treated neurons. Exposing organotypic slices to 30 nM corticosterone reduced apical length in a more delayed manner: only neurons examined more than 2 hours after exposure to corticosterone showed atrophy of the apical dendritic tree. Neither dose of corticosterone affected the length of basal dendrites or spine density. Corticosterone was ineffective in changing morphology of the apical dendrites when tested in the presence of the glucocorticoid receptor antagonist RU38486. These results suggest that high physiological levels of corticosterone, via activation of the glucocorticoid receptor, can, during the course of only a few hours, reduce the dendritic complexity of CA1 pyramidal neurons in young, developing hippocampal tissue. These findings suggest that it is relevant to maintain plasma corticosterone levels low during hippocampal development.
Collapse
Affiliation(s)
- Deborah N Alfarez
- Swammerdam Institute for Life Sciences, Center for Neuroscience, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The impact of stress on brain function is increasingly recognized. Various substances are released in response to stress and can influence distinct neuronal circuits, but the functional advantages of having such a diversity of stress mediators remain unclear. Individual neurotransmitter, neuropeptide and steroid stress mediators have specific spatial and temporal niches, but these niches also overlap. In addition, the effects of individual mediators on neuronal function and plasticity are integrated, and emerging evidence suggests that there is crosstalk between them. Together, this results in the stress instruments producing an orchestrated 'symphony' that enables fine-tuned responses to diverse challenges.
Collapse
Affiliation(s)
- Marian Joëls
- SILS-CNS, University of Amsterdam, the Netherlands.
| | | |
Collapse
|
35
|
Kaku N, Matsuda KI, Tsujimura A, Kawata M. Characterization of nuclear import of the domain-specific androgen receptor in association with the importin alpha/beta and Ran-guanosine 5'-triphosphate systems. Endocrinology 2008; 149:3960-9. [PMID: 18420738 PMCID: PMC2488236 DOI: 10.1210/en.2008-0137] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 04/08/2008] [Indexed: 12/11/2022]
Abstract
Androgen induces androgen receptor (AR) nuclear import, which allows AR to act as a transcriptional factor and ultimately leads to biological activity. However, the mechanism of AR translocation to the nucleus is still unclear. In the present study, we assessed the nuclear import abilities of each domain of AR and their mechanisms related to Ran and importin alpha/beta using green fluorescent protein real-time imaging. The localization of AR to the nucleus in the absence and presence of ligands was dependent upon a complex interplay of the amino terminal transactivation domain (NTD), the DNA binding domain (DBD), and the ligand binding domain (LBD). NTD and DBD showed ligand-independent nuclear import ability, whereas LBD had ligand-dependent transport. In addition, AR deletion mutant lacking DBD was distributed in the cytoplasm regardless of ligand existence, suggesting that the remaining domains, NTD and LBD, are responsible for AR cytoplasmic localization. Cotransfection with a dominant negative form of Ran dramatically inhibited the nuclear import of all AR domains, and a dominant negative form of importin alpha prevented AR and DBD import. Importin beta-knockdown strongly blocked DBD import. These results indicate that there are two additional nuclear localization signals (NLSs) in the NTD and LBD, and there are distinct pathways used to attain domain-specific AR nuclear import: the NLS of DBD is Ran and importin alpha/beta-dependent, whereas the NLSs of NTD and LBD are Ran dependent but importin alpha/beta-independent. Our data suggest that the nuclear import of AR is regulated by the interplay between each domain of the AR.
Collapse
Affiliation(s)
- Natsuko Kaku
- Departments of Anatomy and Neurobiology, Research Institute for Geriatric and Neurobiological Diseases, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | | | | | | |
Collapse
|
36
|
Brouwer JR, Severijnen E, de Jong FH, Hessl D, Hagerman RJ, Oostra BA, Willemsen R. Altered hypothalamus-pituitary-adrenal gland axis regulation in the expanded CGG-repeat mouse model for fragile X-associated tremor/ataxia syndrome. Psychoneuroendocrinology 2008; 33:863-73. [PMID: 18472227 PMCID: PMC4408208 DOI: 10.1016/j.psyneuen.2008.03.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2008] [Revised: 03/19/2008] [Accepted: 03/26/2008] [Indexed: 10/22/2022]
Abstract
The human FMR1 gene contains an unstable CGG-repeat in its 5' untranslated region. The repeat length in the normal population is polymorphic (5-54 CGG-repeats). Individuals carrying lengths beyond 200 CGGs (i.e. the full mutation) show hypermethylation and as a consequence gene silencing of the FMR1 gene. The absence of the gene product FMRP causes the fragile X syndrome, the most common inherited form of mental retardation. Elderly carriers of the premutation (PM), which is defined as a repeat length between 55 and 200 CGGs, can develop a progressive neurodegenerative syndrome: fragile X-associated tremor/ataxia syndrome (FXTAS). The high FMR1 mRNA levels observed in cells from PM carriers have led to the hypothesis that FXTAS is caused by a pathogenic RNA gain-of-function mechanism. Apart from tremor/ataxia, specific psychiatric symptoms have been described in PM carriers with or without FXTAS. Since these symptoms could arise from elevated stress hormone levels, we investigated hypothalamic-pituitary-adrenal (HPA) axis regulation using a knock-in mouse model with an expanded CGG-repeat in the PM range (>98 repeats) in the Fmr1 gene, which shows repeat instability, and displays biochemical, phenotypic and neuropathological characteristics of FXTAS. We show elevated levels of corticosterone in serum and ubiquitin-positive inclusions in both the pituitary and adrenal gland of 100-week-old animals. In addition, we demonstrate ubiquitin-positive inclusions in the amygdala from aged expanded CGG-repeat mice. We hypothesize that altered regulation of the HPA axis and the amygdala and higher stress hormone levels in the mouse model for FXTAS may explain associated psychological symptoms in humans.
Collapse
Affiliation(s)
- JR Brouwer
- Department of Clinical Genetics, Erasmus MC Rotterdam, 3000 CA, The Netherlands
| | - E Severijnen
- Department of Clinical Genetics, Erasmus MC Rotterdam, 3000 CA, The Netherlands
| | - FH de Jong
- Department of Internal Medicine, Erasmus MC Rotterdam, 3000 CA, The Netherlands
| | - D Hessl
- Medical Investigation of Neurodevelopmental Disorders (M.I.N.D.) Institute, School of Medicine, University of California–Davis, Sacramento, CA, 95817 USA
,Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California-Davis, Sacramento, CA, 95817 USA
| | - RJ Hagerman
- Department of Psychiatry and Behavioral Sciences, School of Medicine, University of California-Davis, Sacramento, CA, 95817 USA
,Department of Pediatrics, University of California at Davis School of Medicine, Sacramento, CA, 95817 USA
| | - BA Oostra
- Department of Clinical Genetics, Erasmus MC Rotterdam, 3000 CA, The Netherlands
| | - R Willemsen
- Department of Clinical Genetics, Erasmus MC Rotterdam, 3000 CA, The Netherlands
,Correspondence to: Willemsen R, Department of Clinical Genetics, Erasmus MC, PO Box 2040, 3000 CA Rotterdam, The Netherlands, , TEL: +31 10 7043152, FAX: +31 10 7044736
| |
Collapse
|
37
|
Li W, Yang X, Wang K, Tan W, He Y, Guo Q, Tang H, Liu J. Real-time imaging of protein internalization using aptamer conjugates. Anal Chem 2008; 80:5002-8. [PMID: 18533682 DOI: 10.1021/ac800930q] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Angiogenin is a potent angiogenic factor that is known to play an important role in tumor angiogenesis. In this paper, we investigate the cellular internalization of angiogenin conjugated with its highly specific aptamer. By using fluorophore-labeled aptamer and confocal laser scanning microscopy, we have developed a novel and simple method by which to visualize the real-time process of angiogenin internalization. Specifically, when aptamer-angiogenin conjugates were added into cell cultures, conjugates could be selectively bound to HUVE cells (human umbilical vein endothelial cells) and MCF-7 cells (human breast cancer cells). Nuclear staining and Z-axis scanning studies demonstrated that the aptamer-angiogenin conjugates were internalized to intracellular organelles, and dynamic confocal imaging studies indicated that the conjugates were quickly internalized. These results provide the first evidence that a fluorophore-labeled aptamer can be used as a fluorescent probe to visualize the spatiotemporal process of protein internalization in real time.
Collapse
Affiliation(s)
- Wei Li
- College of Chemistry and Chemical Engineering, Engineering Center for Biomedicine, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, PR China
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kawata M, Nishi M, Matsuda K, Sakamoto H, Kaku N, Masugi-Tokita M, Fujikawa K, Hirahara-Wada Y, Takanami K, Mori H. Steroid receptor signalling in the brain--lessons learned from molecular imaging. J Neuroendocrinol 2008; 20:673-6. [PMID: 18601688 DOI: 10.1111/j.1365-2826.2008.01727.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Studies with green fluorescent protein (GFP) have revealed the subcellular distribution of many steroid hormone receptors to be much more dynamic than previously thought. Fluorescence resonance energy transfer (FRET) and fluorescence recovery after photobleaching (FRAP) are powerful techniques with which to examine protein-protein interaction and the mobility of tagged proteins, respectively. FRET analysis revealed that steroid treatment (with corticosterone or testosterone) induces direct interaction of the glucocorticoid receptor (GR) and importin alpha in the cytoplasm and that, shortly after nuclear entry, the GR detaches from importin alpha. The mineralocorticoid receptor (MR) and androgen receptor (AR) show the same trafficking. Upon oestradiol treatment, ERalpha and ERbeta in the same cell are relocalised to form a discrete pattern and are localised in the same discrete cluster (subnuclear foci). FRAP analysis showed that nuclear ERalpha and ERbeta are most dynamic and mobile in the absence of the ligand, and that mobility decreases slightly after ligand treatment. Genomic as well as non-genomic actions of steroid hormones influence the cellular function of target tissues spacio-temporally.
Collapse
Affiliation(s)
- M Kawata
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kyoto, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Agler M, Prack M, Zhu Y, Kolb J, Nowak K, Ryseck R, Shen D, Cvijic ME, Somerville J, Nadler S, Chen T. A high-content glucocorticoid receptor translocation assay for compound mechanism-of-action evaluation. ACTA ACUST UNITED AC 2007; 12:1029-41. [PMID: 17989426 DOI: 10.1177/1087057107309353] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Ligand-induced cytoplasm to nucleus translocation is a critical event in the nuclear receptor (NR) signal transduction cascade. The development of green fluorescent proteins and their color variants fused with NRs, along with the recent developments in automated cellular imaging technologies, has provided unique tools to monitor and quantify the NR translocation events. These technology developments have important implications in the mechanistic evaluation of NR signaling and provide a powerful tool for drug discovery. The unique challenges for developing a robust NR translocation assay include cytotoxicity accompanied with chronic overexpression of NRs, basal translocation induced by serum present in culture medium, and interference from endogenous NRs, as well as subcellular dynamics. The authors have developed a robust assay system for the glucocorticoid receptor (GR) that was applied to a panel of nuclear receptor ligands. Using a high-content imaging system, ligand-induced, dose-dependent GR nuclear translocation was quantified and a correlation with other conventional assays established.
Collapse
Affiliation(s)
- Michele Agler
- Bristol-Myers Squibb, Lead Discovery & Profiling, Wallingford, Connecticut 06492, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Griekspoor A, Zwart W, Neefjes J, Michalides R. Visualizing the action of steroid hormone receptors in living cells. NUCLEAR RECEPTOR SIGNALING 2007; 5:e003. [PMID: 17464358 PMCID: PMC1853070 DOI: 10.1621/nrs.05003] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Accepted: 02/09/2007] [Indexed: 12/30/2022]
Abstract
Transcription controlled by Steroid Hormone Receptors (SHRs) plays a key role in many important physiological processes like organ development, metabolite homeostasis, and response to external stimuli. Understandably, the members of this family have drawn a lot of attention from the scientific community since their discovery, four decades ago. Still, after many years of research we are only beginning to unravel the complex nature of these receptors. The pace at which we do has improved significantly in recent years with the discovery of genetically encoded fluorescent probes, and the accompanying revival of biophysical approaches that allow more detailed study of SHRs. Here, we will look into the different aspects of SHR signalling, and discuss how biophysical techniques have contributed to visualizing their function in their native context, the living cell.
Collapse
|
41
|
Dames P, Laner A, Maucksch C, Aneja MK, Rudolph C. Targeting of the glucocorticoid hormone receptor with plasmid DNA comprising glucocorticoid response elements improves nonviral gene transfer efficiency in the lungs of mice. J Gene Med 2007; 9:820-9. [PMID: 17668918 DOI: 10.1002/jgm.1082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND It has been previously demonstrated that plasmid DNA transport into the nucleus could be increased by transcription factor binding. We chose the glucocorticoid responsive element (GRE) which binds to the glucocorticoid receptor (GR), a transcription factor which is shuttled into the nucleus upon ligand binding such as dexamethasone. METHODS We cloned two, four, and eight repetitive sequences of the GRE into the reporter plasmid pEGFPLuc. Binding of the pEGFPLuc-GRE to the GR was examined by electrophoretic mobility shift assay (EMSA) experiments. GR expression in bronchiolar and alveolar epithelial cells was confirmed by Western blotting. Intracellular trafficking of GR was examined using a fusion protein of cyano-fluorescent protein (CFP) and GR. Transfection efficiencies of pEGFPLuc compared to pEGFPLucGRE(2-8) were examined in vitro and in vivo upon tail vein injection of cationic liposome gene vectors containing dexamethasone (safeplexes) and aerosol application of polyethylenimine (PEI)-pDNA particles. RESULTS Binding of GRE containing plasmids to the GR was shown in EMSA experiments and intranuclear shuttling of CFP-GR after ligand stimulation was confirmed. Enhanced gene transfer efficiency of pEGFPLucGRE(2) in vitro was only observed on confluent cells. A 2.5-fold increase in gene expression in the lungs of mice after tail vein injection of pEGFPLucGRE(2) complexed with safeplexes compared with pEGFPLuc was observed. PEI-mediated aerosol gene delivery of pEGFPLucGRE(2) was 4.7-fold higher than pEGFPLuc only after intraperitoneal dexamethasone. CONCLUSION The results suggest that inclusion of GRE sequences into plasmid DNA vectors combined with dexamethasone application could improve transgene expression in the lungs in vivo.
Collapse
Affiliation(s)
- Petra Dames
- Department of Pediatrics, Ludwig-Maximilians University, Lindwurmstrasse 2a, 80337, Munich, Germany
| | | | | | | | | |
Collapse
|
42
|
Arnett-Mansfield RL, Graham JD, Hanson AR, Mote PA, Gompel A, Scurr LL, Gava N, de Fazio A, Clarke CL. Focal subnuclear distribution of progesterone receptor is ligand dependent and associated with transcriptional activity. Mol Endocrinol 2006; 21:14-29. [PMID: 17021053 DOI: 10.1210/me.2006-0041] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The progesterone receptor (PR) is a critical mediator of progesterone action in the female reproductive system. Expressed in the human as two proteins, PRA and PRB, the receptor is a ligand-activated nuclear transcription factor that regulates transcription by interaction with protein cofactors and binding to specific response elements in target genes. We previously reported that PR was located in discrete subnuclear foci in human endometrium. In this study, we investigated the role of ligand in the formation of PR foci and their association with transcriptional activity. PR foci were detected in mouse uterus and normal human breast tissues and were more abundant when circulating progesterone was high. In human malignant tissues, PR foci were aberrant: foci were larger in endometrial cancers than in normal endometrium, and in breast cancers hormone-dependence was decreased. Chromatin disruption also increased foci size and decreased ligand dependence, suggesting that altered nuclear architecture may contribute to the aberrant PR foci observed in endometrial and breast cancers. In breast cancer cells, movement of PR into foci required exposure to ligand and was blocked by transcriptional inhibitors and by prolonged inhibition of proteasomal degradation. Foci contained PR dimers, and fluorescence resonance energy transfer demonstrated that PR foci contained the highest concentration of receptor dimers in the nucleus. PR in foci colocalized with transcription factors and nascent RNA transcripts only in the presence of ligand, and inhibition of coactivator recruitment inhibited PR foci formation. The demonstration that focal distribution of PR within the nucleus is associated with transcription suggests a link between the subnuclear distribution of PR and its transcriptional activity that is likely to be important for normal cellular function of PR.
Collapse
|
43
|
Solá S, Amaral JD, Borralho PM, Ramalho RM, Castro RE, Aranha MM, Steer CJ, Rodrigues CMP. Functional Modulation of Nuclear Steroid Receptors by Tauroursodeoxycholic Acid Reduces Amyloid β-Peptide-Induced Apoptosis. Mol Endocrinol 2006; 20:2292-303. [PMID: 16728529 DOI: 10.1210/me.2006-0063] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Tauroursodeoxycholic acid (TUDCA) prevents amyloid beta-peptide (Abeta)-induced neuronal apoptosis, by modulating both classical mitochondrial pathways and specific upstream targets. In addition, activation of nuclear steroid receptors (NSRs), such as the mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) differentially regulates apoptosis in the brain. In this study we investigated whether TUDCA, a cholesterol-derived endogenous molecule, requires NSRs for inhibiting Abeta-induced apoptosis in primary neurons. Our results confirmed that TUDCA significantly reduced Abeta-induced apoptosis; in addition, the fluorescently labeled bile acid molecule was detected diffusely in both cytoplasm and nucleus of rat cortical neurons. Interestingly, experiments using small interfering RNAs (siRNAs) revealed that, in contrast to GR siRNA, MR siRNA abolished the antiapoptotic effect of TUDCA. Abeta incubation reduced MR nuclear translocation while increasing nuclear GR levels. Notably, pretreatment with TUDCA markedly altered Abeta-induced changes in NSRs, including MR dissociation from its cytosolic chaperone, heat shock protein 90, and subsequent translocation to the nucleus. Furthermore, when a carboxy terminus-deleted form of MR was used, nuclear trafficking of both MR and the bile acid was abrogated, suggesting that they translocate to the nucleus as a steroid-receptor complex. Transfection experiments with wild-type or mutant MR confirmed that this interaction was required for TUDCA protection against Abeta-induced apoptosis. Finally, in cotransfection experiments with NSR response element reporter and overexpression constructs, pretreatment with TUDCA significantly modulated Abeta-induced changes in MR and GR transactivation. In conclusion, these results provide novel insights into the specific cellular mechanism of TUDCA antiapoptotic function against Abeta-induced apoptosis and suggest targets for potential therapeutic intervention.
Collapse
Affiliation(s)
- Susana Solá
- Centro de Patogénese Molecular, Faculty of Pharmacy, University of Lisbon, Lisbon 1600-083, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Karst H, Berger S, Turiault M, Tronche F, Schütz G, Joëls M. Mineralocorticoid receptors are indispensable for nongenomic modulation of hippocampal glutamate transmission by corticosterone. Proc Natl Acad Sci U S A 2005; 102:19204-7. [PMID: 16361444 PMCID: PMC1323174 DOI: 10.1073/pnas.0507572102] [Citation(s) in RCA: 574] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The adrenal hormone corticosterone transcriptionally regulates responsive genes in the rodent hippocampus through nuclear mineralocorticoid and glucocorticoid receptors. Via this genomic pathway the hormone alters properties of hippocampal cells slowly and for a prolonged period. Here we report that corticosterone also rapidly and reversibly changes hippocampal signaling. Stress levels of the hormone enhance the frequency of miniature excitatory postsynaptic potentials in CA1 pyramidal neurons and reduce paired-pulse facilitation, pointing to a hormone-dependent enhancement of glutamate-release probability. The rapid effect by corticosterone is accomplished through a nongenomic pathway involving membrane-located receptors. Unexpectedly, the rapid effect critically depends on the classical mineralocorticoid receptor, as evidenced by the effectiveness of agonists, antagonists, and brain-specific inactivation of the mineralocorticoid but not the glucocorticoid receptor gene. Rapid actions by corticosterone would allow the brain to change its function within minutes after stress-induced elevations of corticosteroid levels, in addition to responding later through gene-mediated signaling pathways.
Collapse
Affiliation(s)
- Henk Karst
- Swammerdam Institute for Life Sciences, Center for Neurosciences (SILS-CNS), University of Amsterdam, Kruislaan 320, 1098 SM Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
45
|
Komatsuzaki Y, Murakami G, Tsurugizawa T, Mukai H, Tanabe N, Mitsuhashi K, Kawata M, Kimoto T, Ooishi Y, Kawato S. Rapid spinogenesis of pyramidal neurons induced by activation of glucocorticoid receptors in adult male rat hippocampus. Biochem Biophys Res Commun 2005; 335:1002-7. [PMID: 16111661 DOI: 10.1016/j.bbrc.2005.07.173] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2005] [Accepted: 07/20/2005] [Indexed: 11/18/2022]
Abstract
Modulation of hippocampal synaptic plasticity by glucocorticoids has been attracting much attention, due to its importance in stress responses. Dendritic spines are essential for memory storage processes. Here, we investigated the effect of dexamethasone (DEX), a specific agonist of glucocorticoid receptor (GR), on density and morphology of dendritic spines in adult male rat hippocampus by imaging of Lucifer Yellow-injected spines in slices. The application of 100 nM DEX (stressful high concentration) induced rapid modulation of the density and morphology of dendritic spines in CA1 pyramidal neurons within 1h. The total spine density increased from 0.88 spines/microm (control) to 1.36 spines/microm (DEX-treated). DEX significantly increased the density of thin and mushroom type spines, however only a slight increase was observed for stubby and filopodium type spines. Because the presence of 10 microM cycloheximide, an inhibitor of protein synthesis, did not suppress the DEX effect, these responses are probably non-genomic. Western immunoblot analysis demonstrated the localization of classical type GR in Triton-insoluble synaptosomal fractions (enriched in postsynaptic membranes) from hippocampal slices, suggesting a possible action site of DEX at spines.
Collapse
Affiliation(s)
- Yoshimasa Komatsuzaki
- School of Science and Technology, Meiji University, 1-1-1 Higasimita, Tama, Kawasaki, Kanagawa 214, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Scott GR, Keir KR, Schulte PM. Effects of spironolactone and RU486 on gene expression and cell proliferation after freshwater transfer in the euryhaline killifish. J Comp Physiol B 2005; 175:499-510. [PMID: 16088394 DOI: 10.1007/s00360-005-0014-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2005] [Revised: 04/20/2005] [Accepted: 06/14/2005] [Indexed: 10/25/2022]
Abstract
We have explored the possible mechanisms by which mineralocorticoid (MR) and glucocorticoid (GR) receptors regulate the response to freshwater transfer in the gills of the euryhaline killifish Fundulus heteroclitus. Killifish were implanted with RU486 (GR antagonist) or spironolactone (MR antagonist) at doses of 0.1-1.0 mg g(-1), and subsequently transferred from 10 per thousand brackish water to freshwater. Compared to brackish water sham fish, mRNA expression of CFTR and NKCC1 decreased in the gills of sham fish transferred to freshwater, whereas Na(+), K(+)-ATPase alpha(1a) mRNA expression and alpha protein abundance, as well as cell proliferation (detected using BrdU) increased. Spironolactone inhibited the normal increase in cell proliferation and Na(+), K(+)-ATPase expression after freshwater transfer. RU486 increased plasma cortisol levels and may have slightly inhibited Na(+), K(+)-ATPase activity, but did not change alpha(1a ) expression. RU486 had no effect on cell proliferation in the non-lamellar region of the gills, but increased proliferation in the lamellar region. Neither antagonist inhibited the suppression of CFTR or NKCC1 expression after freshwater transfer. Glucocorticoid receptor expression was reduced in all sham and antagonist treatments compared to untreated controls, but no other consistent differences were observed. The effects of spironolactone suggest that MR is important for regulating ion transport in killifish gills after freshwater transfer.
Collapse
Affiliation(s)
- Graham R Scott
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada.
| | | | | |
Collapse
|
47
|
Abstract
In response to stress, the brain activates several neuropeptide-secreting systems. This eventually leads to the release of adrenal corticosteroid hormones, which subsequently feed back on the brain and bind to two types of nuclear receptor that act as transcriptional regulators. By targeting many genes, corticosteroids function in a binary fashion, and serve as a master switch in the control of neuronal and network responses that underlie behavioural adaptation. In genetically predisposed individuals, an imbalance in this binary control mechanism can introduce a bias towards stress-related brain disease after adverse experiences. New candidate susceptibility genes that serve as markers for the prediction of vulnerable phenotypes are now being identified.
Collapse
Affiliation(s)
- E Ron de Kloet
- Department of Medical Pharmacology, Leiden Amsterdam Center for Drug Research and Leiden University Medical Center, Gorlaeus Laboratories, Leiden University, PO Box 9502, 2300 RA Leiden, The Netherlands.
| | | | | |
Collapse
|
48
|
Post A, Ohl F, Almeida OFX, Binder EB, Rücker M, Welt S, Binder E, Holsboer F, Sillaber I. Identification of molecules potentially involved in mediating the in vivo actions of the corticotropin-releasing hormone receptor 1 antagonist, NBI30775 (R121919). Psychopharmacology (Berl) 2005; 180:150-8. [PMID: 15682302 DOI: 10.1007/s00213-004-2134-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2004] [Accepted: 11/19/2004] [Indexed: 10/25/2022]
Abstract
RATIONALE The neuropeptide corticotropin-releasing hormone (CRH) plays a central role in the regulation of the hypothalamo-pituitary-adrenocortical (HPA) axis. The view that CRH hypersecretion underlies anxiety and mood disorders was recently supported by preclinical and clinical data obtained after application of the CRH receptor (CRH-R1) antagonist NBI30775 (R121919). Despite its therapeutic efficacy, there is only little information about its mechanisms of action on cellular and molecular targets. OBJECTIVE To identify some of the intracellular substrates mediating the actions of NBI30775 after its acute administration in a stress-independent animal model. RESULTS Of the different doses of NBI30775 tested (0.5, 1, 5 and 30 mg/kg), the 1-mg/kg dose proved behaviorally active insofar that it reduced anxiety-like behavior in mice under basal conditions. Subsequent analysis of brain tissues revealed NBI30775-induced increases in the nuclear translocation of glucocorticoid receptors (GR) and BAG-1, an upregulation of mRNA transcripts encoding GR, mineralocorticoid receptors (MR) and CRH-R1, and a suppression of the DNA-binding activity of the transcription factor AP-1. These changes were significant at a dose of 1 mg/kg of NBI30775. CONCLUSION NBI30775 reduces levels of anxiety in mice (under basal conditions) with a steep dose-response curve. Molecules such as GR, MR, BAG-1 and AP-1 have been identified as some of the drug's intracellular targets; interestingly, changes in these molecules have also been seen in response to conventional antidepressants, showing that structurally and mechanistically unrelated anxiolytic and antidepressant drugs can influence common downstream pathways.
Collapse
MESH Headings
- Animals
- Anti-Anxiety Agents/administration & dosage
- Anti-Anxiety Agents/pharmacology
- Anxiety/drug therapy
- Anxiety/metabolism
- Brain/drug effects
- Brain/metabolism
- Corticosterone/blood
- Dose-Response Relationship, Drug
- Gene Expression
- Male
- Mice
- Mice, Inbred DBA
- NF-kappa B/biosynthesis
- NF-kappa B/genetics
- Pyrimidines/administration & dosage
- Pyrimidines/pharmacology
- RNA, Messenger/biosynthesis
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/biosynthesis
- Receptors, Corticotropin-Releasing Hormone/genetics
- Receptors, Glucocorticoid/biosynthesis
- Receptors, Glucocorticoid/genetics
- Receptors, Mineralocorticoid/biosynthesis
- Receptors, Mineralocorticoid/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factor AP-1/biosynthesis
- Transcription Factor AP-1/genetics
- Transcriptional Activation
- Up-Regulation
Collapse
Affiliation(s)
- Anke Post
- Max Planck Institute of Psychiatry, Kraepelinstrasse 10, 80804 Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Pratt WB, Galigniana MD, Harrell JM, DeFranco DB. Role of hsp90 and the hsp90-binding immunophilins in signalling protein movement. Cell Signal 2005; 16:857-72. [PMID: 15157665 DOI: 10.1016/j.cellsig.2004.02.004] [Citation(s) in RCA: 199] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2003] [Accepted: 01/30/2004] [Indexed: 11/16/2022]
Abstract
The ubiquitous protein chaperone hsp90 has been shown to regulate more than 100 proteins involved in cellular signalling. These proteins are called 'client proteins' for hsp90, and a multiprotein hsp90/hsp70-based chaperone machinery forms client protein.hsp90 heterocomplexes in the cytoplasm and the nucleus. In the case of signalling proteins that act as transcription factors, the client protein.hsp90 complexes also contain one of several TPR domain immunophilins or immunophilin homologs that bind to a TPR domain binding site on hsp90. Using several intracellular receptors and the tumor suppressor p53 as examples, we review evidence that dynamic assembly of heterocomplexes with hsp90 is required for rapid movement through the cytoplasm to the nucleus along microtubular tracks. The role of the immunophilin in this system is to connect the client protein.hsp90 complex to cytoplasmic dynein, the motor protein for retrograde movement toward the nucleus. Upon arrival at the nuclear pores, the receptor.hsp90.immunophilin complexes are transferred to the nuclear interior by importin-dependent facilitated diffusion. The unliganded receptors then distribute within the nucleus to diffuse patches from which they proceed in a ligand-dependent manner to discrete nuclear foci where chromatin binding occurs. We review evidence that dynamic assembly of heterocomplexes with hsp90 is required for movement to these foci and for the dynamic exchange of transcription factors between chromatin and the nucleoplasm.
Collapse
Affiliation(s)
- William B Pratt
- Department of Pharmacology, University of Michigan Medical School, 1301 Med. Sci. Res. Building III, Ann Arbor, MI 48109-0632, USA.
| | | | | | | |
Collapse
|
50
|
Tanaka M, Nishi M, Morimoto M, Sugimoto T, Kawata M. Imaging analysis of mineralocorticoid receptor and importins in single living cells by using GFP color variants. Cell Tissue Res 2005; 320:447-53. [PMID: 15846517 DOI: 10.1007/s00441-004-0984-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Accepted: 08/10/2004] [Indexed: 11/27/2022]
Abstract
Mineralocorticoid receptor (MR) is a ligand-dependent transcription factor involved in gene regulation in association with another corticosteroid receptor, glucocorticoid receptor. In the absence of ligand, MR resides both in the cytoplasm and in the nucleus. Agonists increase the number of MRs residing in the nucleus. Importins are docking proteins for karyopherin-mediated binding of substrate in a nuclear import pathway. To investigate the interactions between MR and importins, we analyzed the subcellular distribution of MR and importins in response to ligand in living COS-1 cells, which do not express endogenous MR, by using fusion proteins labeled with different spectral variants of green fluorescent protein. In the cells coexpressing fluorescent protein-tagged (FP)-MR and FP-importin alpha, the proteins simultaneously moved into the nucleus from the cytoplasm upon activation with ligand treatment. In the cells coexpressing FP-MR and FP-importin beta, FP-MR moved into the nucleus from the cytoplasm, but the distribution of FP-importin beta was little changed upon ligand treatment. Analysis of a mutant of MR, in which nuclear localization signal (NLS) is inactivated, demonstrated that the intact NLS is necessary for the trafficking of MR related to importin alpha. This is the first visual evidence of the nuclear import of MR in association with importin alpha in single living cells.
Collapse
Affiliation(s)
- Masayuki Tanaka
- Department of Anatomy and Neurobiology, Kyoto Prefectural University of Medicine, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | | | | | | | | |
Collapse
|