201
|
Jenkins SI, Pickard MR, Khong M, Smith HL, Mann CL, Emes RD, Chari DM. Identifying the cellular targets of drug action in the central nervous system following corticosteroid therapy. ACS Chem Neurosci 2014; 5:51-63. [PMID: 24147833 PMCID: PMC3894723 DOI: 10.1021/cn400167n] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/21/2013] [Indexed: 12/11/2022] Open
Abstract
Corticosteroid (CS) therapy is used widely in the treatment of a range of pathologies, but can delay production of myelin, the insulating sheath around central nervous system nerve fibers. The cellular targets of CS action are not fully understood, that is, "direct" action on cells involved in myelin genesis [oligodendrocytes and their progenitors the oligodendrocyte precursor cells (OPCs)] versus "indirect" action on other neural cells. We evaluated the effects of the widely used CS dexamethasone (DEX) on purified OPCs and oligodendrocytes, employing complementary histological and transcriptional analyses. Histological assessments showed no DEX effects on OPC proliferation or oligodendrocyte genesis/maturation (key processes underpinning myelin genesis). Immunostaining and RT-PCR analyses show that both cell types express glucocorticoid receptor (GR; the target for DEX action), ruling out receptor expression as a causal factor in the lack of DEX-responsiveness. GRs function as ligand-activated transcription factors, so we simultaneously analyzed DEX-induced transcriptional responses using microarray analyses; these substantiated the histological findings, with limited gene expression changes in DEX-treated OPCs and oligodendrocytes. With identical treatment, microglial cells showed profound and global changes post-DEX addition; an unexpected finding was the identification of the transcription factor Olig1, a master regulator of myelination, as a DEX responsive gene in microglia. Our data indicate that CS-induced myelination delays are unlikely to be due to direct drug action on OPCs or oligodendrocytes, and may occur secondary to alterations in other neural cells, such as the immune component. To the best of our knowledge, this is the first comparative molecular and cellular analysis of CS effects in glial cells, to investigate the targets of this major class of anti-inflammatory drugs as a basis for myelination deficits.
Collapse
Affiliation(s)
- Stuart I. Jenkins
- Institute for Science
and Technology in Medicine, School of Medicine, Keele University, David Weatherall building, Keele, Staffordshire ST5
5BG, United Kingdom
| | - Mark R. Pickard
- Institute for Science
and Technology in Medicine, School of Medicine, Keele University, David Weatherall building, Keele, Staffordshire ST5
5BG, United Kingdom
| | - Melinda Khong
- School of Veterinary
Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, United Kingdom
| | - Heather L. Smith
- School of Veterinary
Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, United Kingdom
| | - Carl L.A. Mann
- Neurology Department, University Hospital of North Staffordshire NHS Trust, City General, Newcastle Road, Stoke-on-Trent, Staffordshire ST4 6QG, United Kingdom
| | - Richard D. Emes
- School of Veterinary
Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, United Kingdom
- Advanced Data Analysis Centre, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire LE12 5RD, United Kingdom
| | - Divya M. Chari
- Institute for Science
and Technology in Medicine, School of Medicine, Keele University, David Weatherall building, Keele, Staffordshire ST5
5BG, United Kingdom
| |
Collapse
|
202
|
Treatment of chronic inflammatory demyelinating polyneuropathy: from molecular bases to practical considerations. Autoimmune Dis 2014; 2014:201657. [PMID: 24527207 PMCID: PMC3914592 DOI: 10.1155/2014/201657] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is an autoimmune disease of the peripheral nervous system, in which both cellular and humoral immune responses are involved. The disease is clinically heterogeneous with some patients displaying pure motor form and others also showing a variable degree of sensory dysfunction; disease evolution may also differ from patient to patient, since monophasic, progressive, and relapsing forms are reported. Underlying such clinical variability there is probably a broad spectrum of molecular dysfunctions that are and will be the target of therapeutic strategies. In this review we first explore the biological bases of current treatments and subsequently we focus on the practical management that must also take into account pharmacoeconomic issues.
Collapse
|
203
|
Krieger S, Sorrells SF, Nickerson M, Pace TWW. Mechanistic insights into corticosteroids in multiple sclerosis: war horse or chameleon? Clin Neurol Neurosurg 2014; 119:6-16. [PMID: 24635918 DOI: 10.1016/j.clineuro.2013.12.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 11/19/2013] [Accepted: 12/27/2013] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Relapse management is a crucial component of multiple sclerosis (MS) care. High-dose corticosteroids (CSs) are used to dampen inflammation, which is thought to hasten the recovery of MS relapse. A diversity of mechanisms drive the heterogeneous clinical response to exogenous CSs in patients with MS. Preclinical research is beginning to provide important insights into how CSs work, both in terms of intended and unintended effects. In this article we discuss cellular, systemic, and clinical characteristics that might contribute to intended and unintended CS effects when utilizing supraphysiological doses in clinical practice. The goal of this article is to consider recent insights about CS mechanisms of action in the context of MS. METHODS We reviewed relevant preclinical and clinical studies on the desirable and undesirable effects of high-dose corticosteroids used in MS care. RESULTS Preclinical studies reviewed suggest that corticosteroids may act in unpredictable ways in the context of autoimmune conditions. The precise timing, dosage, duration, cellular exposure, and background CS milieu likely contribute to their clinical heterogeneity. CONCLUSION It is difficult to predict when patients will respond favorably to CSs, both in terms of therapeutic response and tolerability profile. There are specific cellular, systemic, and clinical characteristics that might merit further consideration when utilizing CSs in clinical practice, and these should be explored in a translational setting.
Collapse
Affiliation(s)
- Stephen Krieger
- Corinne Goldsmith Dickinson Center for MS, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shawn F Sorrells
- Department of Neurosurgery, The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, California, USA
| | | | - Thaddeus W W Pace
- College of Nursing and College of Medicine (Department of Psychiatry), University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|
204
|
Carvalho LA, Bergink V, Sumaski L, Wijkhuijs J, Hoogendijk WJ, Birkenhager TK, Drexhage HA. Inflammatory activation is associated with a reduced glucocorticoid receptor alpha/beta expression ratio in monocytes of inpatients with melancholic major depressive disorder. Transl Psychiatry 2014; 4:e344. [PMID: 24424390 PMCID: PMC3905228 DOI: 10.1038/tp.2013.118] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 09/09/2013] [Accepted: 10/29/2013] [Indexed: 01/01/2023] Open
Abstract
In this study, we used new technology to investigate whether a coherent pattern of enhanced expression of inflammatory and other immune activation genes in circulating monocytes is found in patients with major depression. Since a high inflammatory state of monocytes might be related to glucocorticoid resistance, we also included the genes for the two isoforms of the glucocorticoid receptor. For this study, we aimed at finding a similar coherent pattern of inflammatory and immune activation genes in monocytes of patients with MDD and recruited 47 medication-free melancholic MDD inpatients and 42 healthy controls. A quantitative-polymerase chain reaction (Q-PCR) monocyte gene expression analysis was performed using a panel of inflammatory-related genes previously identified as abnormally regulated in mood disorder patients. Selected serum cytokines/chemokines were assessed using a cytometric bead array. Depressive symptoms were analysed using Hamilton depression scores (HAMD). Thirty-four of the 47 monocyte inflammatory-related genes were significantly upregulated and 2 were significantly downregulated as compared to controls, the latter including the gene for the active GRα in particular in those with a high HAMD score. The reduced GRα expression correlated strongly to the upregulation of the inflammatory genes in monocytes. Serum levels of IL6, IL8, CCL2 and VEGF were significantly increased in patients compared to controls. Our data show the deregulation of two interrelated homoeostatic systems, that is, the immune system and the glucocorticoid system, co-occurring in major depression.
Collapse
Affiliation(s)
- L A Carvalho
- Department of Epidemiology and Public Health, University College London, London, UK,UCL Research Department of Epidemiology and Public Health, 1–19 Torrington Place, Rm356a, London, WC1E 7HB, UK. E-mail:
| | - V Bergink
- Department of Psychiatry, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - L Sumaski
- Klinik und Poliklinik fur Psychiatrie und Psychotherapie, Universitatsklinikum Munster, Munster, Germany
| | - J Wijkhuijs
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - W J Hoogendijk
- Department of Psychiatry, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - T K Birkenhager
- Department of Psychiatry, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - H A Drexhage
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
205
|
Li M, Woo PTK. Glucocorticoid receptors on and in a unicellular organism, Cryptobia salmositica. Int J Parasitol 2013; 44:205-10. [PMID: 24333137 DOI: 10.1016/j.ijpara.2013.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 10/26/2013] [Accepted: 10/28/2013] [Indexed: 11/17/2022]
Abstract
This is the first report to our knowledge that demonstrates a functional steroid hormone receptor in a protozoon. The study used Cryptobia salmositica, a pathogenic haemoflagellate found in salmonid fishes. It has been previously shown that cortisol and dexamethasone (a synthetic glucocorticoid) enhanced the multiplication of C. salmositica under in vitro conditions indicating the presence of glucocorticoid receptors on/in the parasite. Also, the glucocorticoid receptor antagonist, mifepristone (RU486), inhibited the stimulatory effect of the two glucocorticoids on parasite multiplication. In the present study, we used an antibody (produced in a rabbit against glucocorticoid receptor protein) agglutination test and confocal microscopy with immunohistofluorescence staining to demonstrate cortisol-glucocorticoid receptor-like protein receptors on the plasma membrane and in the cytoplasm of the parasite. In two in vitro studies, the addition of 50ngml(-1) of RU486 was more effective in inhibiting parasite replication in cultures with 7,000parasitesml(-1) than in cultures with 14,000parasitesml(-1). Also, 100ngml(-1) of RU486/ml was more effective than 50ngml(-1) in inhibiting parasite multiplication in the 14,000 parasitesml(-1) cultures. These in vitro studies indicate that the number of binding sites on/in the parasite is finite. The findings may be important in future studies especially on steroid receptor signalling pathways and dissection of ligand-receptor interactions, and for evaluating the adaptations that develop in pathogens as part of the host-parasite interaction.
Collapse
Affiliation(s)
- Mao Li
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Patrick T K Woo
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
206
|
Revollo JR, Oakley RH, Lu NZ, Kadmiel M, Gandhavadi M, Cidlowski JA. HES1 is a master regulator of glucocorticoid receptor-dependent gene expression. Sci Signal 2013; 6:ra103. [PMID: 24300895 DOI: 10.1126/scisignal.2004389] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Hairy and enhancer of split-1 (HES1) is a basic helix-loop-helix transcription factor that is a key regulator of development and organogenesis. However, little is known about the role of HES1 after birth. Glucocorticoids, primary stress hormones that are essential for life, regulate numerous homeostatic processes that permit vertebrates to cope with physiological challenges. The molecular actions of glucocorticoids are mediated by glucocorticoid receptor-dependent regulation of nearly 25% of the genome. Here, we established a genome-wide molecular link between HES1 and glucocorticoid receptors that controls the ability of cells and animals to respond to stress. Glucocorticoid signaling rapidly and robustly silenced HES1 expression. This glucocorticoid-dependent repression of HES1 was necessary for the glucocorticoid receptor to regulate many of its target genes. Mice with conditional knockout of HES1 in the liver exhibited an expanded glucocorticoid receptor signaling profile and aberrant metabolic phenotype. Our results indicate that HES1 acts as a master repressor, the silencing of which is required for proper glucocorticoid signaling.
Collapse
Affiliation(s)
- Javier R Revollo
- 1Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | |
Collapse
|
207
|
Whirledge S, Cidlowski JA. A role for glucocorticoids in stress-impaired reproduction: beyond the hypothalamus and pituitary. Endocrinology 2013; 154:4450-68. [PMID: 24064362 PMCID: PMC3836069 DOI: 10.1210/en.2013-1652] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In addition to the well-characterized role of the sex steroid receptors in regulating fertility and reproduction, reproductive events are also mediated by the hypothalamic-pituitary-adrenal axis in response to an individual's environment. Glucocorticoid secretion in response to stress contributes to the well-characterized suppression of the hypothalamic-pituitary-gonadal axis through central actions in the hypothalamus and pituitary. However, both animal and in vitro studies indicate that other components of the reproductive system are also regulated by glucocorticoids. Furthermore, in the absence of stress, it appears that homeostatic glucocorticoid signaling plays a significant role in reproduction and fertility in all tissues comprising the hypothalamic-pituitary-gonadal axis. Indeed, as central regulators of the immune response, glucocorticoids are uniquely poised to integrate an individual's infectious, inflammatory, stress, nutritional, and metabolic status through glucocorticoid receptor signaling in target tissues. Endocrine signaling between tissues regulating the immune and stress response and those determining reproductive status provides an evolutionary advantage, facilitating the trade-off between reproductive investment and offspring fitness. This review focuses on the actions of glucocorticoids in tissues important for fertility and reproduction, highlighting recent studies that show glucocorticoid signaling plays a significant role throughout the hypothalamic-pituitary-gonadal axis and characterizing these effects as permissive or inhibitory in terms of facilitating reproductive success.
Collapse
Affiliation(s)
- Shannon Whirledge
- NIEHS/NIH, MD F3-07, P.O. Box 12233, Research Triangle Park, North Carolina 27709.
| | | |
Collapse
|
208
|
Effect of a high-fat--high-fructose diet, stress and cinnamon on central expression of genes related to immune system, hypothalamic-pituitary-adrenocortical axis function and cerebral plasticity in rats. Br J Nutr 2013; 111:1190-201. [PMID: 24252462 DOI: 10.1017/s0007114513003577] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The intake of a high-fat/high-fructose (HF/HFr) diet is described to be deleterious to cognitive performances, possibly via the induction of inflammatory factors. An excess of glucocorticoids is also known to exert negative effects on cerebral plasticity. In the present study, we assessed the effects of an unbalanced diet on circulating and central markers of inflammation and glucocorticoid activity, as well as their reversal by dietary cinnamon (CN) supplementation. A group of male Wistar rats were subjected to an immune challenge with acute lipopolysaccharide under a HF/HFr or a standard diet. Another group of Wistar rats were fed either a HF/HFr or a control diet for 12 weeks, with or without CN supplementation, and with or without restraint stress (Str) application before being killed. We evaluated the effects of such regimens on inflammation parameters in the periphery and brain and on the expression of actors of brain plasticity. To assess hypothalamic-pituitary-adrenocortical axis activity, we measured the plasma concentrations of corticosterone and the expression of central corticotrophin-releasing hormone, mineralocorticoid receptor, glucocorticoid receptor and 11β-hydroxysteroid dehydrogenase. We found that the HF/HFr diet induced the expression of cytokines in the brain, but only after an immune challenge. Furthermore, we observed the negative effects of Str on the plasma concentrations of corticosterone and neuroplasticity markers in rats fed the control diet but not in those fed the HF/HFr diet. Additionally, we found that CN supplementation exerted beneficial effects under the control diet, but that its effects were blunted or even reversed under the HF/HFr diet. CN supplementation could be beneficial under a standard diet. [corrected].
Collapse
|
209
|
Indyk JA, Candido-Vitto C, Wolf IM, Venkataraman S, Munoz R, Saladino RA, Witchel SF, Defranco DB. Reduced glucocorticoid receptor protein expression in children with critical illness. Horm Res Paediatr 2013; 79:169-78. [PMID: 23548248 DOI: 10.1159/000348290] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 01/20/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The diagnostic criteria for critical illness-related corticoid insufficiency (CIRCI) are not well established, particularly for children. In addition to alterations in adrenal function, cellular resistance to glucocorticoid action could contribute to CIRCI due to alterations in the functioning of the intracellular receptor protein for corticosteroids, the glucocorticoid receptor (GR). METHODS We have therefore undertaken a pilot, prospective study to assess whether cellular GR activity can be measured in peripheral blood mononuclear cells (PBMCs) from critically ill children. RESULTS Total and cytoplasmic, but not nuclear GR levels were significantly lower in PBMCs from critically ill children (i.e. sepsis/septic shock and traumatic brain injury) compared to healthy controls . While total cortisol concentrations did not differ between test groups, salivary and serum-free cortisol concentrations were significantly greater in both groups of children with critical illness. Cortisol-binding globulin levels were significantly lower in patients with sepsis/septic shock. CONCLUSIONS The lower total and cytoplasmic receptor levels in critically ill children suggest that the GR-mediated response to exogenous glucocorticoid therapy may be limited. However, the nuclear transport of GR in critically ill patients suggests that residual receptors in these patients retain functionality and may be accessible to therapeutic treatments that maximize their activity.
Collapse
Affiliation(s)
- Justin A Indyk
- Division of Pediatric Endocrinology, Children's Hospital of Pittsburgh of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
| | | | | | | | | | | | | | | |
Collapse
|
210
|
Strehl C, Buttgereit F. Optimized glucocorticoid therapy: teaching old drugs new tricks. Mol Cell Endocrinol 2013; 380:32-40. [PMID: 23403055 DOI: 10.1016/j.mce.2013.01.026] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/31/2013] [Accepted: 01/31/2013] [Indexed: 01/01/2023]
Abstract
Glucocorticoids (GCs) are commonly used in the treatment of a wide range of rheumatic and other inflammatory diseases. They exert their potent anti-inflammatory and immunosuppressive effects primarily via so called genomic mechanisms, mediated by the cytosolic glucocorticoid receptor (cGR). This mechanism of GC action can be divided into the transactivation and the transrepression processes. However, also rapid effects of GCs exist which are mediated by specific and unspecific non-genomic mechanisms. A clinical relevance of this mode of GC action is assumed for effects mediated by membrane-bound glucocorticoid receptors, but detailed knowledge on the underlying mechanisms is still missing. Great efforts have been made in the past to diminish GC-induced adverse effects, thus improving the benefit/risk ratio of the drugs. Besides approaches to improve the treatment with conventional glucocorticoids currently available to clinicians, new innovative GCs or GC receptor ligands are also being developed.
Collapse
Affiliation(s)
- Cindy Strehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany.
| | | |
Collapse
|
211
|
Rose AJ, Herzig S. Metabolic control through glucocorticoid hormones: an update. Mol Cell Endocrinol 2013; 380:65-78. [PMID: 23523966 DOI: 10.1016/j.mce.2013.03.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 02/21/2013] [Accepted: 03/08/2013] [Indexed: 01/28/2023]
Abstract
In the past decades, glucocorticoid (GC) hormones and their cognate, intracellular receptor, the glucocorticoid receptor (GR), have been well established as critical checkpoints in mammalian energy homeostasis. Whereas many aspects in healthy nutrient metabolism require physiological levels and/or action of GC, aberrant GC/GR signalling has been linked to severe metabolic dysfunction, including obesity, insulin resistance and type 2 diabetes. Consequently, studies of the molecular mechanisms within the GC signalling axis have become a major focus in biomedical research, up-to-date particularly focusing on systemic glucose and lipid handling. However, with the availability of novel high throughput technologies and more sophisticated metabolic phenotyping capabilities, as-yet non-appreciated, metabolic functions of GC have been recently discovered, including regulatory roles of the GC/GR axis in protein and bile acid homeostasis as well as metabolic inter-organ communication. Therefore, this review summarises recent advances in GC/GR biology, and summarises findings relevant for basic and translational metabolic research.
Collapse
Affiliation(s)
- Adam J Rose
- Joint Research Division, Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg, Center for Molecular Biology (ZMBH), Heidelberg University, Network Aging Research, University Hospital Heidelberg, Germany
| | | |
Collapse
|
212
|
Ratman D, Vanden Berghe W, Dejager L, Libert C, Tavernier J, Beck IM, De Bosscher K. How glucocorticoid receptors modulate the activity of other transcription factors: a scope beyond tethering. Mol Cell Endocrinol 2013; 380:41-54. [PMID: 23267834 DOI: 10.1016/j.mce.2012.12.014] [Citation(s) in RCA: 289] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/13/2012] [Accepted: 12/16/2012] [Indexed: 01/11/2023]
Abstract
The activity of the glucocorticoid receptor (GR), a nuclear receptor transcription factor belonging to subclass 3C of the steroid/thyroid hormone receptor superfamily, is typically triggered by glucocorticoid hormones. Apart from driving gene transcription via binding onto glucocorticoid response elements in regulatory regions of particular target genes, GR can also inhibit gene expression via transrepression, a mechanism largely based on protein:protein interactions. Hereby GR can influence the activity of other transcription factors, without contacting DNA itself. GR is known to inhibit the activity of a growing list of immune-regulating transcription factors. Hence, GCs still rule the clinic for treatments of inflammatory disorders, notwithstanding concomitant deleterious side effects. Although patience is a virtue when it comes to deciphering the many mechanisms GR uses to influence various signaling pathways, the current review is testimony of the fact that groundbreaking mechanistic work has been accumulating over the past years and steadily continues to grow.
Collapse
Affiliation(s)
- Dariusz Ratman
- Cytokine Receptor Lab, VIB Department of Medical Protein Research, VIB, UGent, Albert Baertsoenkaai 3, B-9000 Gent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
213
|
Heberden C, Meffray E, Goustard-Langelier B, Maximin E, Lavialle M. Dexamethasone inhibits the maturation of newly formed neurons and glia supplemented with polyunsaturated fatty acids. J Steroid Biochem Mol Biol 2013; 138:395-402. [PMID: 23907015 DOI: 10.1016/j.jsbmb.2013.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 06/20/2013] [Accepted: 07/22/2013] [Indexed: 01/04/2023]
Abstract
Stress bears a negative impact on adult neurogenesis. High levels of corticoids have been shown to inhibit neural stem cell proliferation, and are considered responsible for the loss of neural precursors. Their effects on the differentiation of the glial and neuronal lineages have been less studied. We examined the effect of dexamethasone (Dex), a synthetic glucocorticoid, on the differentiation of rat neural stem cells in vitro. Dex had no effect on the differentiation of cells cultured under standard conditions. Since we previously determined that NSC, when cultured under classical conditions, were deprived of polyunsaturated fatty acids (PUFA), and displayed phospholipid compositions very different from the in vivo figures [1], we examined the effect of Dex under PUFA supplementation. Dex impaired neuron and oligodendrocyte maturation in PUFA-supplemented cells, demonstrated by the reduction of neurite lengths and oligodendrocyte sizes. This effect was mediated by the glucocorticoid receptor (GR), since it was eliminated by mifepristone, a GR antagonist, and could be relayed by a reduction of ERK phosphorylation. We determined that GR was associated with PPAR β and α under basal conditions, and that this association was disrupted when PUFA were added in combination with Dex. We assumed that this effect on the receptor status enabled the effect of Dex on PUFA supplemented cells, since we determined that the binding to the glucocorticoid response element was higher in cells incubated with PUFA and Dex. In conclusion, corticoids can impair NSC differentiation, and consequently impact the entire process of neurogenesis.
Collapse
Affiliation(s)
- Christine Heberden
- INRA UR0909 Laboratoire de Nutrition et Régulations Lipidiques des Fonctions Cérébrales, CRJ 78352 Jouy-en-Josas, France.
| | | | | | | | | |
Collapse
|
214
|
Quax RA, Manenschijn L, Koper JW, Hazes JM, Lamberts SWJ, van Rossum EFC, Feelders RA. Glucocorticoid sensitivity in health and disease. Nat Rev Endocrinol 2013; 9:670-86. [PMID: 24080732 DOI: 10.1038/nrendo.2013.183] [Citation(s) in RCA: 209] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids regulate many physiological processes and have an essential role in the systemic response to stress. For example, gene transcription is modulated by the glucocorticoid-glucocorticoid receptor complex via several mechanisms. The ultimate biologic responses to glucocorticoids are determined by not only the concentration of glucocorticoids but also the differences between individuals in glucocorticoid sensitivity, which is influenced by multiple factors. Differences in sensitivity to glucocorticoids in healthy individuals are partly genetically determined by functional polymorphisms of the gene that encodes the glucocorticoid receptor. Hereditary syndromes have also been identified that are associated with increased and decreased sensitivity to glucocorticoids. As a result of their anti-inflammatory properties, glucocorticoids are widely used in the treatment of allergic, inflammatory and haematological disorders. The variety in clinical responses to treatment with glucocorticoids reflects the considerable variation in glucocorticoid sensitivity between individuals. In immune-mediated disorders, proinflammatory cytokines can induce localized resistance to glucocorticoids via several mechanisms. Individual differences in how tissues respond to glucocorticoids might also be involved in the predisposition for and pathogenesis of the metabolic syndrome and mood disorders. In this Review, we summarize the mechanisms that influence glucocorticoid sensitivity in health and disease and discuss possible strategies to modulate glucocorticoid responsiveness.
Collapse
Affiliation(s)
- Rogier A Quax
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 's-Gravendijkwal 230, 3015 CE Rotterdam, Netherlands
| | | | | | | | | | | | | |
Collapse
|
215
|
Cell-intrinsic regulation of murine dendritic cell function and survival by prereceptor amplification of glucocorticoid. Blood 2013; 122:3288-97. [PMID: 24081658 DOI: 10.1182/blood-2013-03-489138] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although the inhibitory effects of therapeutic glucocorticoids (GCs) on dendritic cells (DCs) are well established, the roles of endogenous GCs in DC homeostasis are less clear. A critical element regulating endogenous GC concentrations involves local conversion of inactive substrates to active 11-hydroxyglucocorticoids, a reduction reaction catalyzed within the endoplasmic reticulum by an enzyme complex containing 11β-hydroxysteroid dehydrogenase type 1 (11βHSD1) and hexose-6-phosphate dehydrogenase (H6PDH). In this study, we found that this GC amplification pathway operates both constitutively and maximally in steady state murine DC populations and is unaffected by additional inflammatory stimuli. Under physiologic conditions, 11βHSD1-H6PDH increases the sensitivity of plasmacytoid DCs (pDCs) to GC-induced apoptosis and restricts the survival of this population through a cell-intrinsic mechanism. Upon CpG activation, the effects of enzyme activity are overridden, with pDCs becoming resistant to GCs and fully competent to release type I interferon. CD8α(+) DCs are also highly proficient in amplifying GC levels, leading to impaired maturation following toll-like receptor-mediated signaling. Indeed, pharmacologic inhibition of 11βHSD1 synergized with CpG to enhance specific T-cell responses following vaccination targeted to CD8α(+) DCs. In conclusion, amplification of endogenous GCs is a critical cell-autonomous mechanism for regulating the survival and functions of DCs in vivo.
Collapse
|
216
|
Essential role of stress hormone signaling in cardiomyocytes for the prevention of heart disease. Proc Natl Acad Sci U S A 2013; 110:17035-40. [PMID: 24082121 DOI: 10.1073/pnas.1302546110] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Heart failure is a leading cause of death in humans, and stress is increasingly associated with adverse cardiac outcomes. Glucocorticoids are primary stress hormones, but their direct role in cardiovascular health and disease is poorly understood. To determine the in vivo function of glucocorticoid signaling in the heart, we generated mice with cardiomyocyte-specific deletion of the glucocorticoid receptor (GR). These mice are born at the expected Mendelian ratio, but die prematurely from spontaneous cardiovascular disease. By 3 mo of age, mice deficient in cardiomyocyte GR display a marked reduction in left ventricular systolic function, as evidenced by decreases in ejection fraction and fractional shortening. Heart weight and left ventricular mass are elevated, and histology revealed cardiac hypertrophy without fibrosis. Removal of endogenous glucocorticoids and mineralocorticoids neither augmented nor lessened the hypertrophic response. Global gene expression analysis of knockout hearts before pathology onset revealed aberrant regulation of a large cohort of genes associated with cardiovascular disease as well as unique disease genes associated with inflammatory processes. Genes important for maintaining cardiac contractility, repressing cardiac hypertrophy, promoting cardiomyocyte survival, and inhibiting inflammation had decreased expression in the GR-deficient hearts. These findings demonstrate that a deficiency in cardiomyocyte glucocorticoid signaling leads to spontaneous cardiac hypertrophy, heart failure, and death, revealing an obligate role for GR in maintaining normal cardiovascular function. Moreover, our findings suggest that selective activation of cardiomyocyte GR may represent an approach for the prevention of heart disease.
Collapse
|
217
|
The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol 2013; 132:1033-44. [PMID: 24084075 DOI: 10.1016/j.jaci.2013.09.007] [Citation(s) in RCA: 711] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 12/20/2022]
Abstract
Glucocorticoids are primary stress hormones necessary for life that regulate numerous physiologic processes in an effort to maintain homeostasis. Synthetic derivatives of these hormones have been mainstays in the clinic for treating inflammatory diseases, autoimmune disorders, and hematologic cancers. The physiologic and pharmacologic actions of glucocorticoids are mediated by the glucocorticoid receptor (GR), a member of the nuclear receptor superfamily of ligand-dependent transcription factors. Ligand-occupied GR induces or represses the transcription of thousands of genes through direct binding to DNA response elements, physically associating with other transcription factors, or both. The traditional view that glucocorticoids act through a single GR protein has changed dramatically with the discovery of a large cohort of receptor isoforms with unique expression, gene-regulatory, and functional profiles. These GR subtypes are derived from a single gene by means of alternative splicing and alternative translation initiation mechanisms. Posttranslational modification of these GR isoforms further expands the diversity of glucocorticoid responses. Here we discuss the origin and molecular properties of the GR isoforms and their contribution to the specificity and sensitivity of glucocorticoid signaling in healthy and diseased tissues.
Collapse
|
218
|
Kadmiel M, Cidlowski JA. Glucocorticoid receptor signaling in health and disease. Trends Pharmacol Sci 2013; 34:518-30. [PMID: 23953592 PMCID: PMC3951203 DOI: 10.1016/j.tips.2013.07.003] [Citation(s) in RCA: 589] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/30/2013] [Accepted: 07/08/2013] [Indexed: 12/20/2022]
Abstract
Glucocorticoids are steroid hormones regulated in a circadian and stress-associated manner to maintain various metabolic and homeostatic functions that are necessary for life. Synthetic glucocorticoids are widely prescribed drugs for many conditions including asthma, chronic obstructive pulmonary disease (COPD), and inflammatory disorders of the eye. Research in the past few years has begun to unravel the profound complexity of glucocorticoid signaling and has contributed remarkably to improved therapeutic strategies. Glucocorticoids signal through the glucocorticoid receptor (GR), a member of the superfamily of nuclear receptors, in both genomic and non-genomic ways in almost every tissue in the human body. In this review, we provide an update on glucocorticoid receptor signaling and highlight the role of GR signaling in physiological and pathophysiological conditions in the major organ systems in the human body.
Collapse
Affiliation(s)
- Mahita Kadmiel
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T.W. Alexander Dr., MD F3-07, Research Triangle Park, NC 27709
| | - John A. Cidlowski
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T.W. Alexander Dr., MD F3-07, Research Triangle Park, NC 27709
| |
Collapse
|
219
|
Chen KC, Blalock EM, Curran-Rauhut MA, Kadish I, Blalock SJ, Brewer L, Porter NM, Landfield PW. Glucocorticoid-dependent hippocampal transcriptome in male rats: pathway-specific alterations with aging. Endocrinology 2013; 154:2807-20. [PMID: 23736296 PMCID: PMC3713214 DOI: 10.1210/en.2013-1139] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Although glucocorticoids (GCs) are known to exert numerous effects in the hippocampus, their chronic regulatory functions remain poorly understood. Moreover, evidence is inconsistent regarding the long-standing hypothesis that chronic GC exposure promotes brain aging/Alzheimer disease. Here, we adrenalectomized male F344 rats at 15 months of age, maintained them for 3 months with implanted corticosterone (CORT) pellets producing low or intermediate (glucocorticoid receptor-activating) blood levels of CORT, and performed microarray/pathway analyses in hippocampal CA1. We defined the chronic GC-dependent transcriptome as 393 genes that exhibited differential expression between intermediate and low CORT groups. Short-term CORT (4 days) did not recapitulate this transcriptome. Functional processes/pathways overrepresented by chronic CORT-up-regulated genes included learning/plasticity, differentiation, glucose metabolism, and cholesterol biosynthesis, whereas processes overrepresented by CORT-down-regulated genes included inflammatory/immune/glial responses and extracellular structure. These profiles indicate that GCs chronically activate neuronal/metabolic processes while coordinately repressing a glial axis of reactivity/inflammation. We then compared the GC transcriptome with a previously defined hippocampal aging transcriptome, revealing a high proportion of common genes. Although CORT and aging moved expression of some common genes in the same direction, the majority were shifted in opposite directions by CORT and aging (eg, glial inflammatory genes down-regulated by CORT are up-regulated with aging). These results contradict the hypothesis that GCs simply promote brain aging and also suggest that the opposite direction shifts during aging reflect resistance to CORT regulation. Therefore, we propose a new model in which aging-related GC resistance develops in some target pathways, whereas GC overstimulation develops in others, together generating much of the brain aging phenotype.
Collapse
Affiliation(s)
- Kuey-Chu Chen
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
220
|
Yin Y, Zhang X, Li Z, Deng L, Jiao G, Zhang B, Xie P, Mu H, Qiao W, Zou J. Glucocorticoid receptor β regulates injury-mediated astrocyte activation and contributes to glioma pathogenesis via modulation of β-catenin/TCF transcriptional activity. Neurobiol Dis 2013; 59:165-76. [PMID: 23906498 DOI: 10.1016/j.nbd.2013.07.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 07/03/2013] [Accepted: 07/17/2013] [Indexed: 11/16/2022] Open
Abstract
Astrocytes react to central nervous system (CNS) injury and participate in gliotic responses, imparting negative, as well as positive effects on axonal regeneration. Despite the considerable biochemical and morphological changes astrocytes undergo following insult, and the known influence of steroids on glial activation, details surrounding glucocorticoid receptor expression and activity are lacking. Such mechanistic information is essential for advancing and enhancing therapies in the treatment of CNS injuries. Using an in vitro wound-healing assay, we found glucocorticoid receptor β (GRβ), not GRα, is upregulated and acts as a regulator of gliosis after injury. In addition, our results suggest that GRβ interacts with β-catenin and is a necessary component for proliferation and migration in both injured astrocytes and glioma cells. Further analysis indicated GRβ/β-catenin interaction as a key modulator of astrocyte reactivity through sustained Wnt/β-catenin/TCF signaling in its dominant-negative effect on GRα mediated trans-repression by a GSK-3β-independent manner. These findings expand our knowledge of the mechanism of GRβ action in promoting astrocyte proliferation and migration following injury and in glioma. This information furthers our understanding the function of glucocorticoid receptor in CNS injury and disease, as well as in the basic biochemical responses astrocytes undergo in response to injury and glioma pathogenesis.
Collapse
Affiliation(s)
- Ying Yin
- Department of Clinical Laboratory Science, Wuxi People's Hospital of Nanjing Medical University, Wuxi, PR China; Wuxi Clinical Science Research Institute, Wuxi, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Opposing effects of dexamethasone, agrin and sugammadex on functional innervation and constitutive secretion of IL-6 in in vitro innervated primary human muscle cells. Neurosci Lett 2013; 549:186-90. [PMID: 23791923 DOI: 10.1016/j.neulet.2013.06.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 05/28/2013] [Accepted: 06/05/2013] [Indexed: 11/24/2022]
Abstract
Neuromuscular junction development is the key process required for successful neuromuscular transmission and functional innervation of skeletal muscle fibres. Various substances can influence these processes, some of which are in common use in clinical practice. In the present study, the effects of the potentially new therapeutic agent agrin were followed, along with the widely used glucocorticoid dexamethasone. The in vitro experimental model used was functional innervation and constitutive interleukin 6 (IL-6) secretion of human muscle cells. Additionally, the selective relaxant binding agent sugammadex and its possible interaction with dexamethasone were followed. Dexamethasone impaired functional innervation while agrin had opposing effects. Furthermore, based on interference with IL-6 secretion, we show potential (chemical) interactions between dexamethasone and sugammadex. The physiological effects of this interaction should be taken into consideration under clinical conditions where these two drugs might be applied simultaneously.
Collapse
|
222
|
McKeon A. Immunotherapeutics for Autoimmune Encephalopathies and Dementias. Curr Treat Options Neurol 2013; 15:723-37. [DOI: 10.1007/s11940-013-0251-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
223
|
Lee MJ, Pramyothin P, Karastergiou K, Fried SK. Deconstructing the roles of glucocorticoids in adipose tissue biology and the development of central obesity. Biochim Biophys Acta Mol Basis Dis 2013; 1842:473-81. [PMID: 23735216 DOI: 10.1016/j.bbadis.2013.05.029] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Revised: 05/18/2013] [Accepted: 05/24/2013] [Indexed: 12/31/2022]
Abstract
Central obesity is associated with insulin resistance and dyslipidemia. Thus, the mechanisms that control fat distribution and its impact on systemic metabolism have importance for understanding the risk for diabetes and cardiovascular disease. Hypercortisolemia at the systemic (Cushing's syndrome) or local levels (due to adipose-specific overproduction via 11β-hydroxysteroid dehydrogenase 1) results in the preferential expansion of central, especially visceral fat depots. At the same time, peripheral subcutaneous depots can become depleted. The biochemical and molecular mechanisms underlying the depot-specific actions of glucocorticoids (GCs) on adipose tissue function remain poorly understood. GCs exert pleiotropic effects on adipocyte metabolic, endocrine and immune functions, and dampen adipose tissue inflammation. GCs also regulate multiple steps in the process of adipogenesis. Acting synergistically with insulin, GCs increase the expression of numerous genes involved in fat deposition. Variable effects of GC on lipolysis are reported, and GC can improve or impair insulin action depending on the experimental conditions. Thus, the net effect of GC on fat storage appears to depend on the physiologic context. The preferential effects of GC on visceral adipose tissue have been linked to higher cortisol production and glucocorticoid receptor expression, but the molecular details of the depot-dependent actions of GCs are only beginning to be understood. In addition, increasing evidence underlines the importance of circadian variations in GCs in relationship to the timing of meals for determining their anabolic actions on the adipocyte. In summary, although the molecular mechanisms remain to be fully elucidated, there is increasing evidence that GCs have multiple, depot-dependent effects on adipocyte gene expression and metabolism that promote central fat deposition. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
Affiliation(s)
- Mi-Jeong Lee
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, MA, USA
| | - Pornpoj Pramyothin
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, MA, USA; Department of Medicine, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kalypso Karastergiou
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, MA, USA
| | - Susan K Fried
- Department of Medicine, Section of Endocrinology, Diabetes and Nutrition, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
224
|
Lee SR, Kim HK, Song IS, Youm J, Dizon LA, Jeong SH, Ko TH, Heo HJ, Ko KS, Rhee BD, Kim N, Han J. Glucocorticoids and their receptors: insights into specific roles in mitochondria. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 112:44-54. [PMID: 23603102 DOI: 10.1016/j.pbiomolbio.2013.04.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/03/2013] [Accepted: 04/08/2013] [Indexed: 12/27/2022]
Abstract
Glucocorticoids (GCs) affect most physiological systems and are the most frequently used drugs for multiple disorders and organ transplantation. GC functions depend on a balance between circulating GC and cytoplasmic glucocorticoid receptor II (GR). Mitochondria individually enclose circular, double-stranded DNA that is expressed and replicated in response to nuclear-encoded factors imported from the cytoplasm. Fine-tuning and response to cellular demands should be coordinately regulated by the nucleus and mitochondria; thus mitochondrial-nuclear interaction is vital to optimal mitochondrial function. Elucidation of the direct and indirect effects of steroids, including GCs, on mitochondria is an important and emerging field of research. Mitochondria may also be under GC control because GRs are present in mitochondria, and glucocorticoid response elements (GREs) reside in the mitochondrial genome. Therefore, mitochondrial gene expression can be regulated by GCs via at least two different mechanisms: direct action on mitochondrial DNA and oxidative phosphorylation (OXPHOS) genes, or by an indirect effect through interaction with nuclear genes. In this review, we outline possible mechanisms of regulation of mitochondrial genes in response to GCs in view of translocation of the GR into mitochondria and the possible regulation of OXPHOS genes by GREs in the mitochondrial genome.
Collapse
Affiliation(s)
- Sung-Ryul Lee
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, 633-165 Gaegeum-Dong, Busanjin-Gu, 613-735 Busan, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Hu A, Josephson MB, Diener BL, Nino G, Xu S, Paranjape C, Orange JS, Grunstein MM. Pro-asthmatic cytokines regulate unliganded and ligand-dependent glucocorticoid receptor signaling in airway smooth muscle. PLoS One 2013; 8:e60452. [PMID: 23593222 PMCID: PMC3617099 DOI: 10.1371/journal.pone.0060452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 02/26/2013] [Indexed: 01/13/2023] Open
Abstract
To elucidate the regulation of glucocorticoid receptor (GR) signaling under pro-asthmatic conditions, cultured human airway smooth muscle (HASM) cells were treated with proinflammatory cytokines or GR ligands alone and in combination, and then examined for induced changes in ligand-dependent and -independent GR activation and downstream signaling events. Ligand stimulation with either cortisone or dexamethsone (DEX) acutely elicited GR translocation to the nucleus and, comparably, ligand-independent stimulation either with the Th2 cytokine, IL-13, or the pleiotropic cytokine combination, IL-1β/TNFα, also acutely evoked GR translocation. The latter response was potentiated by combined exposure of cells to GR ligand and cytokine. Similarly, treatment with either DEX or IL-13 alone induced GR phosphorylation at its serine-211 residue (GRSer211), denoting its activated state, and combined treatment with DEX+IL-13 elicited heightened and sustained GRSer211 phosphorylation. Interestingly, the above ligand-independent GR responses to IL-13 alone were not associated with downstream GR binding to its consensus DNA sequence or GR transactivation, whereas both DEX-induced GR:DNA binding and transcriptional activity were significantly heightened in the presence of IL-13, coupled to increased recruitment of the transcriptional co-factor, MED14. The stimulated GR signaling responses to DEX were prevented in IL-13-exposed cells wherein GRSer211 phosphorylation was suppressed either by transfection with specific serine phosphorylation-deficient mutant GRs or treatment with inhibitors of the MAPKs, ERK1/2 and JNK. Collectively, these novel data highlight a heretofore-unidentified homeostatic mechanism in HASM cells that involves pro-asthmatic cytokine-driven, MAPK-mediated, non-ligand-dependent GR activation that confers heightened glucocorticoid ligand-stimulated GR signaling. These findings raise the consideration that perturbations in this homeostatic cytokine-driven GR signaling mechanism may be responsible, at least in part, for the insensirtivity to glucocorticoid therapy that is commonly seen in individuals with severe asthma.
Collapse
Affiliation(s)
- Aihua Hu
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Maureen B. Josephson
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Barry L. Diener
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Gustavo Nino
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Integrative Systems Biology and Division of Pulmonary & Sleep Medicine, Children's National Medical Center, Center for Genetic Medicine Research, George Washington University, Washington, D.C., United States of America
| | - Shuyun Xu
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Chinmay Paranjape
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jordan S. Orange
- Section of Immunology, Allergy and Rheumatology, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael M. Grunstein
- Division of Pulmonary Medicine, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania Perlman School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
226
|
Stanczyk FZ, Hapgood JP, Winer S, Mishell DR. Progestogens used in postmenopausal hormone therapy: differences in their pharmacological properties, intracellular actions, and clinical effects. Endocr Rev 2013; 34:171-208. [PMID: 23238854 PMCID: PMC3610676 DOI: 10.1210/er.2012-1008] [Citation(s) in RCA: 306] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The safety of progestogens as a class has come under increased scrutiny after the publication of data from the Women's Health Initiative trial, particularly with respect to breast cancer and cardiovascular disease risk, despite the fact that only one progestogen, medroxyprogesterone acetate, was used in this study. Inconsistency in nomenclature has also caused confusion between synthetic progestogens, defined here by the term progestin, and natural progesterone. Although all progestogens by definition have progestational activity, they also have a divergent range of other properties that can translate to very different clinical effects. Endometrial protection is the primary reason for prescribing a progestogen concomitantly with postmenopausal estrogen therapy in women with a uterus, but several progestogens are known to have a range of other potentially beneficial effects, for example on the nervous and cardiovascular systems. Because women remain suspicious of the progestogen component of postmenopausal hormone therapy in the light of the Women's Health Initiative trial, practitioners should not ignore the potential benefits to their patients of some progestogens by considering them to be a single pharmacological class. There is a lack of understanding of the differences between progestins and progesterone and between individual progestins differing in their effects on the cardiovascular and nervous systems, the breast, and bone. This review elucidates the differences between the substantial number of individual progestogens employed in postmenopausal hormone therapy, including both progestins and progesterone. We conclude that these differences in chemical structure, metabolism, pharmacokinetics, affinity, potency, and efficacy via steroid receptors, intracellular action, and biological and clinical effects confirm the absence of a class effect of progestogens.
Collapse
Affiliation(s)
- Frank Z Stanczyk
- Department of Obstetrics and Gynecology, University of Southern California Keck School of Medicine, Livingston Research Building, 1321 North Mission Road, Room 201, Los Angeles, California 90033, USA.
| | | | | | | |
Collapse
|
227
|
Rovaris DL, Mota NR, de Azeredo LA, Cupertino RB, Bertuzzi GP, Polina ER, Contini V, Kortmann GL, Vitola ES, Grevet EH, Grassi-Oliveira R, Callegari-Jacques SM, Bau CHD. MR and GR functional SNPs may modulate tobacco smoking susceptibility. J Neural Transm (Vienna) 2013; 120:1499-505. [PMID: 23543128 DOI: 10.1007/s00702-013-1012-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 03/15/2013] [Indexed: 10/27/2022]
Abstract
A number of studies have demonstrated that stress is involved in all aspects of smoking behavior, including initiation, maintenance and relapse. The mineralocorticoid (MR) and glucocorticoid (GR) receptors are expressed in several brain areas and play a key role in negative feedback of the hypothalamic-pituitary-adrenal (HPA) axis. As nicotine increases the activation of the HPA axis, we wondered if functional SNPs (single nucleotide polymorphisms) in MR and GR coding genes (NR3C2 rs5522 and NR3C1 rs6198, respectively) may be involved in smoking susceptibility. The sample included 627 volunteers, of which 514 were never-smokers and 113 lifetime smokers. We report an interaction effect between rs5522 and rs6198 SNPs. The odds ratio (OR) for the presence of the NR3C2 rs5522 Val allele in NR3C1 rs6198 G carriers was 0.18 (P = 0.007), while in rs6198 G noncarriers the OR was 1.83 (P = 0.027). We also found main effects of the NR3C1 rs6198 G allele on number of cigarettes smoked per day (P = 0.027) and in total score of the Fagerström Test for Nicotine Dependence (P = 0.007). These findings are consistent with a possible link between NR3C2 and NR3C1 polymorphisms and smoking behavior and provide a first partial replication for a nominally significant GWAS finding between NR3C2 and tobacco smoking.
Collapse
Affiliation(s)
- Diego L Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves - 15053, Porto Alegre, RS, 91501-970, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
The influence of glucocorticoid signaling on tumor progression. Brain Behav Immun 2013; 30 Suppl:S26-31. [PMID: 23164950 PMCID: PMC3987853 DOI: 10.1016/j.bbi.2012.10.022] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 10/22/2012] [Accepted: 10/26/2012] [Indexed: 02/07/2023] Open
Abstract
The diagnosis of cancer elicits a broad range of well-characterized stress-related biobehavioral responses. Recent studies also suggest that an individual's neuroendocrine stress response can influence tumor biology. One of the major physiological pathways altered by the response to unrelenting social stressors is the hypothalamic-pituitary-adrenal or HPA axis. Initially following acute stress exposure, an increased glucocorticoid response is observed; eventually, chronic stress exposure can lead to a blunting of the normal diurnal cortisol pattern. Interestingly, recent evidence also links high primary tumor glucocorticoid receptor expression (and associated increased glucocorticoid-mediated gene expression) to more rapid estrogen-independent breast cancer progression. Furthermore, animal models of human breast cancer suggest that glucocorticoids inhibit tumor cell apoptosis. These findings provide a conceptual basis for understanding the molecular mechanisms underlying the influence of the individual's stress response, and specifically glucocorticoid action, on breast cancer and other solid tumor biology. How this increased glucocorticoid signaling might contribute to cancer progression is the subject of this review.
Collapse
|
229
|
Busillo JM, Cidlowski JA. The five Rs of glucocorticoid action during inflammation: ready, reinforce, repress, resolve, and restore. Trends Endocrinol Metab 2013; 24:109-19. [PMID: 23312823 PMCID: PMC3667973 DOI: 10.1016/j.tem.2012.11.005] [Citation(s) in RCA: 230] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 11/14/2012] [Accepted: 11/20/2012] [Indexed: 02/08/2023]
Abstract
Glucocorticoids are essential for maintaining homeostasis and regulate a wide variety of physiological processes. Therapeutically, synthetic glucocorticoids are widely prescribed for the treatment of inflammation, autoimmune disorders, and malignancies of lymphoid origin. In this review we examine emerging evidence highlighting both proinflammatory and anti-inflammatory actions of glucocorticoids on both the innate and adaptive immune systems. We incorporate these findings into the more traditional anti-inflammatory role attributed to glucocorticoids, and propose how the two seemingly disparate processes seamlessly work together to resolve cellular responses to inflammatory stimuli. These ideas provide a framework by which glucocorticoids ready and reinforce the innate immune system, and repress the adaptive immune system, to help to resolve inflammation and restore homeostasis.
Collapse
Affiliation(s)
- John M Busillo
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
230
|
Ammit AJ. Glucocorticoid insensitivity as a source of drug targets for respiratory disease. Curr Opin Pharmacol 2013; 13:370-6. [PMID: 23434363 DOI: 10.1016/j.coph.2013.02.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 01/24/2013] [Accepted: 02/04/2013] [Indexed: 11/25/2022]
Abstract
Glucocorticoids (corticosteroids) are effective and clinically useful medicines for repressing inflammation in lung disease; however, the number of respiratory conditions that have been recognized to be refractory or insensitive to glucocorticoids is on the rise--either due to an inherent difference in the glucocorticoid sensitivity as part of the disease process or due to exogenous stressors such as cigarette smoke and other oxidative insults. Independent of causality, the aim of future therapeutic advances to conquer this frontier will no doubt be based on our growing knowledge of molecular mechanisms underlying glucocorticoid insensitivity in respiratory diseases. The current article aims to highlight the key molecular mechanisms responsible for glucocorticoid insensitivity in asthma and COPD. This new knowledge will ultimately allow us to enhance lung health by restoring glucocorticoid responsiveness in respiratory disease. In this way, our increased understanding of corticosteroid insensitivity can be exploited as a source of drug targets for respiratory disease in the future.
Collapse
Affiliation(s)
- Alaina J Ammit
- Faculty of Pharmacy, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
231
|
DuBois DC, Sukumaran S, Jusko WJ, Almon RR. Evidence for a glucocorticoid receptor beta splice variant in the rat and its physiological regulation in liver. Steroids 2013; 78:312-20. [PMID: 23257260 PMCID: PMC3552070 DOI: 10.1016/j.steroids.2012.11.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 10/22/2012] [Accepted: 11/22/2012] [Indexed: 12/23/2022]
Abstract
Glucocorticoids are important regulators of metabolism and immune function. Synthetic glucocorticoids are extensively used for immunosuppression/anti-inflammatory therapy. Since the glucocorticoid receptor (GR) is central to most hormone effects; its in vivo regulation will influence hormone/drug action. An alternative splice variant, GRβ, is present in humans and may function as a dominant negative regulator of GR transcriptional activity. Recently, a similar splice variant was reported in mouse, although the mechanism of alternative splicing differs from that in humans. We present evidence that a splice variant of GR with an alternative C-terminus also occurs in the rat by a mechanism of intron inclusion. A highly quantitative qRT-PCR assay for the simultaneous measurement of both splice variants in a single sample was developed in order to accurately measure their regulation. We used this assay to assess the tissue specific expression of both mRNAs, and demonstrate that GRα is predominant in all tissues. In addition, the regulation of both GRα and GRβ mRNA by various physiological factors in rat liver was assessed. GRα showed a robust circadian rhythm, which was entrained with the circadian oscillation of the endogenous hormone. Time series experiments showed that both corticosteroids and LPS but not insulin dosing resulted in the transient down-regulation of GRα mRNA. LPS treatment also resulted in down-regulation of GRβ expression. A modest up-regulation in GRβ expression was observed only in animals having chronically elevated plasma insulin concentrations. However the expression of GRβ was significantly lower than that of GRα in all cases.
Collapse
Affiliation(s)
- Debra C DuBois
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY 14260, USA.
| | | | | | | |
Collapse
|
232
|
Ramamoorthy S, Cidlowski JA. Exploring the molecular mechanisms of glucocorticoid receptor action from sensitivity to resistance. ENDOCRINE DEVELOPMENT 2013; 24:41-56. [PMID: 23392094 PMCID: PMC4770453 DOI: 10.1159/000342502] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Glucocorticoids regulate a variety of physiological processes, and are commonly used to treat disorders of inflammation, autoimmune diseases, and cancer. Glucocorticoid action is predominantly mediated through the classic glucocorticoid receptor (GR), but sensitivity to glucocorticoids varies among individuals, and even within different tissues from the same individual. The molecular basis of this phenomenon can be partially explained through understanding the process of generating bioavailable ligand and the molecular heterogeneity of the GR. The molecular mechanisms that regulate glucocorticoid action highlight the dynamic nature of hormone signaling and provide novel insights into genomic glucocorticoid actions and glucocorticoid sensitivity. Although glucocorticoids are highly effective for therapeutic purposes, long-term and/or high-dose glucocorticoid administration often leads to reduced glucocorticoid sensitivity or resistance. Here, we summarize our current understanding of the mechanisms that modulate glucocorticoid sensitivity and resistance with a focus on GR-mediated signaling.
Collapse
Affiliation(s)
- Sivapriya Ramamoorthy
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, N.C., USA
| | | |
Collapse
|
233
|
Li M, Leatherland JF, Woo PTK. Cortisol and dexamethasone increase the in vitro multiplication of the haemoflagellate, Cryptobia salmositica, possibly by interaction with a glucocorticoid receptor-like protein. Int J Parasitol 2012; 43:353-60. [PMID: 23262305 DOI: 10.1016/j.ijpara.2012.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/19/2012] [Accepted: 11/21/2012] [Indexed: 11/17/2022]
Abstract
Cryptobia salmositica is a pathogenic haemoflagellate of Pacific salmon, Oncorhynchus spp., on the west coast of North America. The in vitro multiplication of the parasite was significantly enhanced by the addition of cortisol (within a range consistent with physiological levels in salmonid fishes; 10-50 ng ml(-1)) to the culture medium (MEM supplemented with FBS). However, higher cortisol concentrations (100 and 200 ng ml(-1)) either had no enhancing effects or resulted in lower replication rates compared with the controls. The synthetic glucocorticoid, dexamethasone (Dex), also stimulated the replication of the parasite and mifepristone (RU486), a synthetic steroid that has glucocorticoid receptor (GR) antagonist properties, inhibited the stimulatory actions of both cortisol and Dex, when added to the medium at a concentration of 100 ng ml(-1) co-culture with cortisol or Dex. Furthermore, the dose-dependent effects of glucocorticoids (cortisol and Dex) on the multiplication of the haemoflagellate were correlated with the initial size of the inocula. The study revealed a novel relationship between the parasite and its host, in which the host's cortisol is used by the parasite to enhance its replication. Also, since C. salmositica responds to both native and synthetic glucocorticoids and to the GR antagonist, RU486, and exhibits a biphasic (hormetic) response to the amount of cortisol in the medium, we propose that the glucocorticoid exerts its effects via an interaction with GR-like proteins in C. salmositica that are functionally similar to those present in vertebrate cells.
Collapse
Affiliation(s)
- Mao Li
- Department of Integrative Biology, University of Guelph, Guelph, ON, Canada
| | | | | |
Collapse
|
234
|
Affiliation(s)
- Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO 80206, UDA.
| |
Collapse
|
235
|
Glucocorticoids induce CCN5/WISP-2 expression and attenuate invasion in oestrogen receptor-negative human breast cancer cells. Biochem J 2012; 447:71-9. [PMID: 22765757 DOI: 10.1042/bj20120311] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CCN5 (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed 5)/WISP-2 [WNT1 (wingless-type MMTV integration site family, member 1)-inducible signalling pathway protein 2] is an oestrogen-regulated member of the CCN family. CCN5 is a transcriptional repressor of genes associated with the EMT (epithelial-mesenchymal transition) and plays an important role in maintenance of the differentiated phenotype in ER (oestrogen receptor)-positive breast cancer cells. In contrast, CCN5 is undetectable in more aggressive ER-negative breast cancer cells. We now report that CCN5 is induced in ER-negative breast cancer cells such as MDA-MB-231 following glucocorticoid exposure, due to interaction of the endogenous glucocorticoid receptor with a functional glucocorticoid-response element in the CCN5 gene promoter. Glucocorticoid treatment of MDA-MB-231 cells is accompanied by morphological alterations, decreased invasiveness and attenuated expression of mesenchymal markers, including vimentin, cadherin 11 and ZEB1 (zinc finger E-box binding homeobox 1). Interestingly, glucocorticoid exposure did not increase CCN5 expression in ER-positive breast cancer cells, but rather down-regulated ER expression, thereby attenuating oestrogen pathway signalling. Taken together, our results indicate that glucocorticoid treatment of ER-negative breast cancer cells induces high levels of CCN5 expression and is accompanied by the appearance of a more differentiated and less invasive epithelial phenotype. These findings propose a novel therapeutic strategy for high-risk breast cancer patients.
Collapse
|
236
|
Ren R, Oakley RH, Cruz-Topete D, Cidlowski JA. Dual role for glucocorticoids in cardiomyocyte hypertrophy and apoptosis. Endocrinology 2012; 153:5346-60. [PMID: 22989630 PMCID: PMC3473206 DOI: 10.1210/en.2012-1563] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Glucocorticoids and their synthetic derivatives are known to alter cardiac function in vivo; however, the nature of these effects and whether glucocorticoids act directly on cardiomyocytes are poorly understood. To explore the role of glucocorticoid signaling in the heart, we used rat embryonic H9C2 cardiomyocytes and primary cardiomyocytes as model systems. Dexamethasone (100 nm) treatment of cardiomyocytes caused a significant increase in cell size and up-regulated the expression of cardiac hypertrophic markers, including atrial natriuretic factor, β-myosin heavy chain, and skeletal muscle α-actin. In contrast, serum deprivation and TNFα exposure triggered cardiomyocyte apoptosis, and these apoptotic effects were inhibited by dexamethasone. Both the hypertrophic and anti-apoptotic actions of glucocorticoids were abolished by the glucocorticoid receptor (GR) antagonist RU486 and by short hairpin RNA-mediated GR depletion. Blocking the activity of the mineralocorticoid receptor had no effect on these glucocorticoid-dependent cardiomyocyte responses. Aldosterone (1 μm) activation of GR also promoted cardiomyocyte hypertrophy and cell survival. To elucidate the mechanism of the dual glucocorticoid actions, a genome-wide microarray was performed on H9C2 cardiomyocytes treated with vehicle or dexamethasone in the absence or presence of serum. Serum dramatically influenced the transcriptome regulated by GR, revealing potential glucocorticoid signaling mediators in both cardiomyocyte hypertrophy and apoptosis. These studies reveal a direct and dynamic role for glucocorticoids and GR signaling in the modulation of cardiomyocyte function.
Collapse
Affiliation(s)
- Rongqin Ren
- Molecular Endocrinology Group, Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, USA
| | | | | | | |
Collapse
|
237
|
Charmandari E, Achermann JC, Carel JC, Soder O, Chrousos GP. Stress response and child health. Sci Signal 2012; 5:mr1. [PMID: 23112343 DOI: 10.1126/scisignal.2003595] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Stress is defined as a state of real or perceived threat to homeostasis. The principal effectors of the stress system are corticotropin-releasing hormone (CRH), arginine vasopressin, the proopiomelanocortin-derived peptides α-melanocyte-stimulating hormone and β-endorphin, the glucocorticoids, and the catecholamines norepinephrine and epinephrine. Appropriate responsiveness of the stress system to stressors is a crucial prerequisite for a sense of well-being, adequate performance of tasks, and positive social interactions. By contrast, inappropriate responsiveness of the stress system may impair growth and development and may account for various endocrine, metabolic, autoimmune, and psychiatric disorders. The development and severity of these conditions primarily depend on the genetic vulnerability of the individual, the exposure to adverse environmental factors, and the timing of stressful event(s). Prenatal life, infancy, childhood, and adolescence are critical periods characterized by increased vulnerability to stressors. This review summarizes the topics presented at the fifth New Inroads to Child Health (NICHe) Conference "Stress Response and Child Health" held at Heraklion, Crete, Greece, in May 2012.
Collapse
Affiliation(s)
- Evangelia Charmandari
- Division of Endocrinology, Metabolism, and Diabetes, First Department of Pediatrics, University of Athens Medical School, Aghia Sophia Children's Hospital, Division of Endocrinology and Metabolism, Athens, Greece.
| | | | | | | | | |
Collapse
|
238
|
Bellavance MA, Rivest S. The neuroendocrine control of the innate immune system in health and brain diseases. Immunol Rev 2012; 248:36-55. [PMID: 22725953 DOI: 10.1111/j.1600-065x.2012.01129.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The innate immune reaction takes place in the brain during immunogenic challenges, injury, and disease. Such a response is highly regulated by numerous anti-inflammatory mechanisms that may directly affect the ultimate consequences of such a reaction within the cerebral environment. The neuroendocrine control of this innate immune system by glucocorticoids is critical for the delicate balance between cell survival and damage in the presence of inflammatory mediators. Glucocorticoids play key roles in regulating the expression of inflammatory genes, and they also have the ability to modulate numerous functions that may ultimately lead to brain damage or repair after injury. Here we review these mechanisms and discuss data supporting both neuroprotective and detrimental roles of the neuroendocrine control of innate immunity.
Collapse
Affiliation(s)
- Marc-André Bellavance
- Laboratory of Endocrinology and Genomics, CHUQ Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | | |
Collapse
|
239
|
Glucocorticoids regulate arrestin gene expression and redirect the signaling profile of G protein-coupled receptors. Proc Natl Acad Sci U S A 2012; 109:17591-6. [PMID: 23045642 DOI: 10.1073/pnas.1209411109] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs) compose the largest family of cell surface receptors and are the most common target of therapeutic drugs. The nonvisual arrestins, β-arrestin-1 and β-arrestin-2, are multifunctional scaffolding proteins that play critical roles in GPCR signaling. On binding of activated GPCRs at the plasma membrane, β-arrestins terminate G protein-dependent responses (desensitization) and stimulate β-arrestin-dependent signaling pathways. Alterations in the cellular complement of β-arrestin-1 and β-arrestin-2 occur in many human diseases, and their genetic ablation in mice has severe consequences. Surprisingly, however, the factors that control β-arrestin gene expression are poorly understood. We demonstrate that glucocorticoids differentially regulate β-arrestin-1 and β-arrestin-2 gene expression in multiple cell types. Glucocorticoids act via the glucocorticoid receptor (GR) to induce the synthesis of β-arrestin-1 and repress the expression of β-arrestin-2. Glucocorticoid-dependent regulation involves the recruitment of ligand-activated glucocorticoid receptors to conserved and functional glucocorticoid response elements in intron-1 of the β-arrestin-1 gene and intron-11 of the β-arrestin-2 gene. In human lung adenocarcinoma cells, the increased expression of β-arrestin-1 after glucocorticoid treatment impairs G protein-dependent activation of inositol phosphate signaling while enhancing β-arrestin-1-dependent stimulation of the MAPK pathway by protease activated receptor 1. These studies demonstrate that glucocorticoids redirect the signaling profile of GPCRs via alterations in β-arrestin gene expression, revealing a paradigm for cross-talk between nuclear and cell surface receptors and a mechanism by which glucocorticoids alter the clinical efficacy of GPCR-based drugs.
Collapse
|
240
|
Kochetov AV, Merkulova TI, Merkulov VM. Possible link between the synthesis of GR alpha isoforms and eIF2 alpha phosphorylation state. Med Hypotheses 2012; 79:709-12. [PMID: 22981593 DOI: 10.1016/j.mehy.2012.07.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/23/2012] [Indexed: 01/30/2023]
Abstract
Glucocorticoid hormones regulate numerous physiological processes and are widely used in the treatment of inflammation, autoimmune disease and cancer. Glucocorticoid receptor (GR) - a transcription factor, derived from a single gene, is responsible for the diverse actions of glucocorticoids. It was shown that GR gene gives rise a variety of mRNA species that produces several protein isoforms, among them GRα is the most abundant. In addition, GRα N-end-truncated protein isoforms (A, B, C, D) are generated by translational mechanisms. As it was found that the ratio between the translational isoforms amounts varied in different tissues and cell lines and distinct isoforms could control transcription of different sets of genes, molecular mechanisms underlining the synthesis of translational GRα isoforms are of great interest. It was considered that GRα isoform A is translated by a conventional linear scanning, isoform B is translated by leaky scanning, isoform C is translated by leaky scanning and ribosomal shunt whereas translation of isoform D occurs through ribosomal shunt only. Since the sequence organization of GRα mRNA strongly resembles the cases of ATF4 or ATF5, the well-known examples of reinitiation-dependent synthesis of functional isoforms, we hypothesize that translation of isoform C could be controlled by reinitiation mechanism also. If this assumption is correct, the ratio between GRα N-end isoforms could depend on the eIF2α phosphorylation state that could provide an additional connection between the GR and cellular stresses. We believe that this hypothesis could be of interest to plan more robust experiments or for better interpretation of available data.
Collapse
|
241
|
Laberge RM, Zhou L, Sarantos MR, Rodier F, Freund A, de Keizer PL, Liu S, Demaria M, Cong YS, Kapahi P, Desprez PY, Hughes RE, Campisi J. Glucocorticoids suppress selected components of the senescence-associated secretory phenotype. Aging Cell 2012; 11:569-78. [PMID: 22404905 PMCID: PMC3387333 DOI: 10.1111/j.1474-9726.2012.00818.x] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cellular senescence suppresses cancer by arresting the proliferation of cells at risk for malignant transformation. Recently, senescent cells were shown to secrete numerous cytokines, growth factors, and proteases that can alter the tissue microenvironment and may promote age-related pathology. To identify small molecules that suppress the senescence-associated secretory phenotype (SASP), we developed a screening protocol using normal human fibroblasts and a library of compounds that are approved for human use. Among the promising library constituents was the glucocorticoid corticosterone. Both corticosterone and the related glucocorticoid cortisol decreased the production and secretion of selected SASP components, including several pro-inflammatory cytokines. Importantly, the glucocorticoids suppressed the SASP without reverting the tumor suppressive growth arrest and were efficacious whether cells were induced to senesce by ionizing radiation or strong mitogenic signals delivered by oncogenic RAS or MAP kinase kinase 6 overexpression. Suppression of the prototypical SASP component IL-6 required the glucocorticoid receptor, which, in the presence of ligand, inhibited IL-1α signaling and NF-κB transactivation activity. Accordingly, co-treatments combining glucocorticoids with the glucocorticoid antagonist RU-486 or recombinant IL-1α efficiently reestablished NF-κB transcriptional activity and IL-6 secretion. Our findings demonstrate feasibility of screening for compounds that inhibit the effects of senescent cells. They further show that glucocorticoids inhibit selected components of the SASP and suggest that corticosterone and cortisol, two FDA-approved drugs, might exert their effects in part by suppressing senescence-associated inflammation.
Collapse
Affiliation(s)
- Remi-Martin Laberge
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Lili Zhou
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China 100875
| | - Melissa R. Sarantos
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM)/Institut du Cancer de Montréal, Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montréal, QC H2L 4M1, Canada
| | - Adam Freund
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Peter L.J. de Keizer
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Su Liu
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Marco Demaria
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Yu-Sheng Cong
- Institute of Cell Biology, College of Life Sciences, Beijing Normal University, Beijing, China 100875
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- California Pacific Medical Center, Research Institute, 475 Brannan Street, San Francisco, CA 94107, USA
| | - Robert E. Hughes
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- Lawrence Berkeley National Laboratory, Life Sciences Division, 1 Cyclotron Road, Berkeley, CA 94720, USA
| |
Collapse
|
242
|
Ligr M, Li Y, Logan SK, Taneja S, Melamed J, Lepor H, Garabedian MJ, Lee P. Mifepristone inhibits GRβ coupled prostate cancer cell proliferation. J Urol 2012; 188:981-8. [PMID: 22819113 DOI: 10.1016/j.juro.2012.04.102] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Indexed: 10/28/2022]
Abstract
PURPOSE The GR gene produces GRα and GRβ isoforms by alternative splicing of a C-terminal exon. GRα binds glucocorticoids, modulates transcription in a glucocorticoid dependent manner and has a growth inhibitory role in prostate cells. Due to this role glucocorticoids are often used to treat androgen independent prostate cancer. In contrast, GRβ has intrinsic transcriptional activity and binds mifepristone (RU486) but not glucocorticoids to control gene expression. To our knowledge the role of GRβ in prostate cell proliferation is unknown. MATERIALS AND METHODS We determined GRβ levels in various prostate cancer cell lines by reverse transcriptase-polymerase chain reaction and Western blot. The effect of GRβ on the kinetics of prostate cancer cell growth was determined by cell counting and flow cytometry upon mifepristone and dexamethasone treatment. Cell proliferation was also examined after siRNA mediated knockdown and over expression of GRβ. RESULTS GRβ mRNA and protein were up-regulated in LNCaP cells that over expressed the androgen receptor co-factor ARA70β. Treatment of LNCaP-ARA70β with mifepristone or siRNA targeting GRβ inhibited proliferation compared to that of parental LNCaP cells. The immortal but nontumorigenic RC165 prostate cell line and the tumorigenic DU145 prostate cell line with endogenous GRβ also showed partial growth reduction upon GRβ depletion but to a lesser extent than LNCaP-ARA70β cells. The growth stimulatory effect of ARA70β on LNCaP cells was partly GRβ dependent, as was the proliferation of RC165 cells and to a lesser extent of DU145 cells. CONCLUSIONS Results suggest that patients with a primary tumor that expresses GRβ and ARA70β may benefit from mifepristone.
Collapse
Affiliation(s)
- Martin Ligr
- Department of Pathology, New York University School of Medicine, New York, New York 10010, USA
| | | | | | | | | | | | | | | |
Collapse
|
243
|
Rubio-Patiño C, Palmeri CM, Pérez-Perarnau A, Cosialls AM, Moncunill-Massaguer C, González-Gironès DM, Pons-Hernández L, López JM, Ventura F, Gil J, Pons G, Iglesias-Serret D. Glycogen synthase kinase-3β is involved in ligand-dependent activation of transcription and cellular localization of the glucocorticoid receptor. Mol Endocrinol 2012; 26:1508-20. [PMID: 22771494 DOI: 10.1210/me.2011-1366] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glucocorticoids (GC) induce cell cycle arrest and apoptosis in different cell types and therefore are widely used to treat a variety of diseases including autoimmune disorders and cancer. This effect is mediated by the GC receptor (GR), a ligand-activated transcription factor that translocates into the nucleus where it modulates transcription of target genes in a promoter-specific manner. Glycogen synthase kinase-3 (GSK3) regulates GR response by genomic and nongenomic mechanisms, although the specific role of each isoform is not well defined. We used GSK3 pharmacological inhibitors and isoform-specific small interfering RNA to evaluate the role of GSK3 in the genomic regulation induced by GC. GSK3 inhibition resulted in the reduction of GC-induced mRNA expression of GC-induced genes such as BIM, HIAP1, and GILZ. Knockdown of GSK3β but not GSK3α reduced endogenous GILZ induction in response to dexamethasone and GR-dependent reporter gene activity. Chromatin immunoprecipitation experiments revealed that GSK3 inhibition impaired the dexamethasone-mediated binding of GR and RNA polymerase II to endogenous GILZ promoter. These results indicate that GSK3β is important for GR transactivation activity and that GSK3β inhibition suppresses GC-stimulated gene expression. Furthermore, we show that genomic regulation by the GR is independent of known GSK3β phosphorylation sites. We propose that GC-dependent transcriptional activation requires functional GSK3β signaling and that altered GSK3β activity influences cell response to GC.
Collapse
Affiliation(s)
- Camila Rubio-Patiño
- Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Merkulov VM, Merkulova TI. Glucocorticoid receptor isoforms generated by alternative splicing and alternative translation initiation. ACTA ACUST UNITED AC 2012. [DOI: 10.1134/s2079059712030070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
245
|
Mittelstadt PR, Monteiro JP, Ashwell JD. Thymocyte responsiveness to endogenous glucocorticoids is required for immunological fitness. J Clin Invest 2012; 122:2384-94. [PMID: 22653054 DOI: 10.1172/jci63067] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/18/2012] [Indexed: 12/21/2022] Open
Abstract
Generation of a self-tolerant but antigen-responsive T cell repertoire occurs in the thymus. Although glucocorticoids are usually considered immunosuppressive, there is also evidence that they play a positive role in thymocyte selection. To address the question of how endogenous glucocorticoids might influence the adaptive immune response, we generated GRlck-Cre mice, in which the glucocorticoid receptor gene (GR) is deleted in thymocytes prior to selection. These mice were immunocompromised, with reduced polyclonal T cell proliferative responses to alloantigen, defined peptide antigens, and viral infection. This was not due to an intrinsic proliferation defect, because GR-deficient T cells responded normally when the TCR was cross-linked with antibodies or when the T cell repertoire was "fixed" with αβ TCR transgenes. Varying the affinity of self ligands in αβ TCR transgenic mice showed that affinities that would normally lead to thymocyte-positive selection caused negative selection, and alterations in the TCR repertoire of polyclonal T cells were confirmed by analysis of TCR Vβ CDR3 regions. Thus, endogenous glucocorticoids are required for a robust adaptive immune response because of their promotion of the selection of T cells that have sufficient affinity for self, and the absence of thymocyte glucocorticoid signaling results in an immunocompromised state.
Collapse
Affiliation(s)
- Paul R Mittelstadt
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | | |
Collapse
|
246
|
The Stat3/GR interaction code: predictive value of direct/indirect DNA recruitment for transcription outcome. Mol Cell 2012; 47:38-49. [PMID: 22633955 DOI: 10.1016/j.molcel.2012.04.021] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Revised: 03/28/2012] [Accepted: 04/16/2012] [Indexed: 11/20/2022]
Abstract
Transcription factor recruitment to genomic sites of action is primarily due to direct protein:DNA interactions. The subsequent recruitment of coregulatory complexes leads to either transcriptional activation or repression. In contrast to this canonical scheme, some transcription factors, such as the glucocorticoid receptor (GR), behave as transcriptional repressors when recruited to target genes through protein tethering. We have investigated the genome-wide prevalence of tethering between GR and Stat3 and found nonreciprocal interactions, namely that GR tethering to DNA-bound Stat3 results in transcriptional repression, whereas Stat3 tethering to GR results in synergism. Further, other schemes of GR and Stat3 corecruitment to regulatory modules result in transcriptional synergism, including neighboring and composite binding sites. The results indicate extensive transcriptional interactions between Stat3 and GR; further, they provide a genome-wide assessment of transcriptional regulation by tethering and a molecular basis for integration of signals mediated by GR and Stats in health and disease.
Collapse
|
247
|
Dibas A, Jiang M, Fudala R, Gryczynski I, Gryczynski Z, Clark AF, Yorio T. Fluorescent protein-labeled glucocorticoid receptor alpha isoform trafficking in cultured human trabecular meshwork cells. Invest Ophthalmol Vis Sci 2012; 53:2938-50. [PMID: 22447868 PMCID: PMC3376074 DOI: 10.1167/iovs.11-8331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 01/17/2012] [Accepted: 03/14/2012] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To characterize the roles of the cytoskeleton and heat shock protein 90 (HSP90) in steroid-induced glucocorticoid receptor alpha (GRα) translocation in cultured human trabecular meshwork cells. METHODS Stably transfected red fluorescent protein (RFP)-GRα NTM5 cell lines were developed. Nuclear localization of RFP-GRα in NTM5 cells treated with vehicle (ethanol), dexamethasone (DEX), or RU486 was measured in cytosolic and nuclear fractions by western blotting and laser confocal microscopy. Cytochalasin D, colchicine, and 17-demethoxygeldanamycin (17AAG, an HSP90 inhibitor), were tested for their abilities to affect GRα trafficking. Nuclear export of RFP-GRα was studied using confocal microscopy following DEX or RU486 removal. RESULTS NTM5 cells transfected with RFP-GRα showed a clear cytosolic localization of receptor that underwent nuclear localization after DEX treatment. RFP-GRα translocation was temperature sensitive, occurring at 37°C but not at room temperature. Neither cytochalasin D nor colchicine blocked DEX-induced or RU486-induced RFP-GRα nuclear translocation; however, 17AAG prevented DEX-induced RFP-GRα nuclear translocation. Both nuclear import and export of DEX-induced RFP-GRα were faster than RU-486-induced nuclear shuttling. CONCLUSIONS RFP-GRα receptor behaves similarly to the wild-type GRα with its cytosolic localization and shuttling to nucleus after DEX or RU486 treatment. HSP90 is required for nuclear translocation, but the disruption of cytoskeleton had no effect on nuclear translocation of RFP-GRα.
Collapse
Affiliation(s)
- Adnan Dibas
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Boulevard, Fort Worth, TX 76107, USA.
| | | | | | | | | | | | | |
Collapse
|
248
|
O'Connor MF, Wellisch DK, Stanton AL, Olmstead R, Irwin MR. Diurnal cortisol in Complicated and Non-Complicated Grief: slope differences across the day. Psychoneuroendocrinology 2012; 37:725-8. [PMID: 21925795 PMCID: PMC3258306 DOI: 10.1016/j.psyneuen.2011.08.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 07/26/2011] [Accepted: 08/23/2011] [Indexed: 11/18/2022]
Abstract
Although grief has been described primarily as a psychological phenomenon, empirical evidence reveals that grief also has physiological correlates that have consequences for health. The present study investigates the diurnal cortisol production patterns in women who have been bereaved in the past 18 months. Specifically, the study compares women with Complicated Grief (n=12) from those with Non-Complicated Grief (n=12), testing whether cortisol slope distinguishes the two groups. Results demonstrate that the two groups do not differ on demographic variables (except education), but as hypothesized, those with Complicated Grief have a flatter slope across the day, controlling for education and body mass index.
Collapse
Affiliation(s)
- Mary-Frances O'Connor
- Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA, United States.
| | | | | | | | | |
Collapse
|
249
|
Chronic restraint stress attenuates p53 function and promotes tumorigenesis. Proc Natl Acad Sci U S A 2012; 109:7013-8. [PMID: 22509031 DOI: 10.1073/pnas.1203930109] [Citation(s) in RCA: 153] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Epidemiological studies strongly suggest that chronic psychological stress promotes tumorigenesis. However, its direct link in vivo and the underlying mechanisms that cause this remain unclear. This study provides direct evidence that chronic stress promotes tumorigenesis in vivo; chronic restraint, a well-established mouse model to induce chronic stress, greatly promotes ionizing radiation (IR)-induced tumorigenesis in p53(+/-) mice. The tumor suppressor protein p53 plays a central role in tumor prevention. Loss or attenuation of p53 function contriubutes greatly to tumorigenesis. We found that chronic restraint decreases the levels and function of p53 in mice, and furthermore, promotes the growth of human xenograft tumors in a largely p53-dependent manner. Our results show that glucocorticoids elevated during chronic restraint mediate the effect of chronic restraint on p53 through the induction of serum- and glucocorticoid-induced protein kinase (SGK1), which in turn increases MDM2 activity and decreases p53 function. Taken together, this study demonstrates that chronic stress promotes tumorigenesis in mice, and the attenuation of p53 function is an important part of the underlying mechanism, which can be mediated by glucocortcoids elevated during chronic restraint.
Collapse
|
250
|
Kumar R, McEwan IJ. Allosteric modulators of steroid hormone receptors: structural dynamics and gene regulation. Endocr Rev 2012; 33:271-99. [PMID: 22433123 PMCID: PMC3596562 DOI: 10.1210/er.2011-1033] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Steroid hormones are synthesized from cholesterol primarily in the adrenal gland and the gonads and play vital roles in normal physiology, the control of development, differentiation, metabolic homeostasis, and reproduction. The actions of these small lipophilic molecules are mediated by intracellular receptor proteins. It is just over 25 yr since the first cDNA for steroid receptors were cloned, a development that led to the birth of a superfamily of ligand-activated transcription factors: the nuclear receptors. The receptor proteins share structurally and functionally related ligand binding and DNA-binding domains but possess distinct N-terminal domains and hinge regions that are intrinsically disordered. Since the original cloning experiments, considerable progress has been made in our understanding of the structure, mechanisms of action, and biology of this important class of ligand-activated transcription factors. In recent years, there has been interest in the structural plasticity and function of the N-terminal domain of steroid hormone receptors and in the allosteric regulation of protein folding and function in response to hormone, DNA response element architecture, and coregulatory protein binding partners. The N-terminal domain can exist as an ensemble of conformers, having more or less structure, which prime this region of the receptor to rapidly respond to changes in the intracellular environment through hormone binding and posttranslation modifications. In this review, we address the question of receptor structure and function dynamics with particular emphasis on the structurally flexible N-terminal domain, intra- and interdomain communications, and the allosteric regulation of receptor action.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Basic Sciences, The Commonwealth Medical College, Scranton, Pennsylvania 18510, USA
| | | |
Collapse
|