301
|
Brunelli E, Pinton G, Chianale F, Graziani A, Appendino G, Moro L. 8-Prenylnaringenin inhibits epidermal growth factor-induced MCF-7 breast cancer cell proliferation by targeting phosphatidylinositol-3-OH kinase activity. J Steroid Biochem Mol Biol 2009; 113:163-70. [PMID: 19103290 DOI: 10.1016/j.jsbmb.2008.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 11/20/2008] [Accepted: 11/25/2008] [Indexed: 01/03/2023]
Abstract
8-Prenylnaringenin (8PN), one of the strongest plant-derived oestrogen receptors (ERs) ligand, has been suggested to have potential cancer chemo-preventive activities and anti-angiogenic properties. Because published data suggest that ERs serve as nodal point that allows interactions between hormones and growth factors mediated pathways, we decided to investigate the effects exerted by 8PN on Epidermal growth factor (EGF)-elicited pathways in breast cancer cells. Here we show that in ER positive MCF-7 cells, 8PN interferes with EGF induced cell proliferation by strongly inhibiting activation of PI(3)K/Akt pathway, without affecting EGFR expression or tyrosine phosphorylation, and exerting a synergistic activation of Erk1/2 phosphorylation. Moreover, we demonstrate that 8PN is a direct inhibitor of PI(3)K activity as it is shown by in vitro experiments with the purified enzyme and by its inability to impair serine phosphorylation of a constitutive active form of Akt. These findings suggest that inhibition of PI(3)K is a novel mechanism which contributes to 8PN activity to inhibit cancer cell survival and EGF induced proliferation.
Collapse
Affiliation(s)
- Elisa Brunelli
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche and Drug and Food Biotechnology Center, Università degli Studi del Piemonte Orientale Amedeo Avogadro, Via Bovio 6, Novara, Italy
| | | | | | | | | | | |
Collapse
|
302
|
Pecci A, Alvarez LD, Veleiro AS, Ceballos NR, Lantos CP, Burton G. New lead compounds in the search for pure antiglucocorticoids and the dissociation of antiglucocorticoid effects. J Steroid Biochem Mol Biol 2009; 113:155-62. [PMID: 19167494 DOI: 10.1016/j.jsbmb.2008.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2008] [Revised: 12/18/2008] [Accepted: 12/22/2008] [Indexed: 11/23/2022]
Abstract
Antiglucocorticoids that act as antagonists at the glucocorticoid receptor (GR) level may be used to block or modulate the undesirable effects of glucocorticoid excess (from endogenous or exogenous origin). RU486 developed in the early 80s, is an antiglucocorticoid but also a potent antiprogestin and abortifacient, nevertheless it still remains as the only GR antagonist drug in the market. Further on, in view of the variety of physiological processes in which glucocorticoids are involved, selective antiglucocorticoids that can block only some of these processes (eventually with tissue specificity) would be highly desirable. The bridged pregnane 21-hydroxy-6,19-epoxyprogesterone, was developed as an alternative lead being an antagonist of the GR with no affinity for mineralocorticoid and progesterone receptors. Antagonistic activity was evidenced by partial blocking of dexamethasone induction of tyrosine aminotransferase (TAT) and thymocyte apoptosis. Replacement of the oxygen bridge by a sulfur bridge gave a less bent, more flexible molecule. 21-Hydroxy-6,19-epithioprogesterone exhibited improved antiapoptotic activity on thymocytes but was not effective blocking TAT induction. This selectivity was improved further by oxidation to the sulfone. The sulfone but not the reduced compound also reverted the dexamethasone-mediated inhibition of NFkappaB activity in HeLa cells. Blocking of the apoptotic effect of TNFalpha by dexamethasone in the L929 cell line (mouse fibroblasts), was only reverted partially by the sulfone which exhibited a mild agonistic/antagonistic activity in this assay. None of these compounds showed antiprogestin activity. Similar overall molecular shapes but more lipophylic and with higher metabolic stability were obtained by introduction of a methylene bridge (6,19-methanoprogesterone) or by a direct bond between C-6 and C-19 (6,19-cycloprogesterone and its 21-hydroxy derivative). The latter highly bent steroids showed affinity for the GR. Recently we performed molecular dynamics simulations of GR-ligand complexes to investigate the molecular basis of the passive antagonism exhibited by 21-hydroxy-6,19-epoxyprogesterone. On the basis of our findings, we proposed that the passive antagonist mode of action of this antiglucocorticoid analog resides, at least in part, in the incapacity of GR-21-hydroxy-6,19-epoxyprogesterone complex to dimerize, making the complex unable to activate gene transcription.
Collapse
Affiliation(s)
- Adali Pecci
- Departamento de Química Biológica (IFIBYNE-CONICET), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
303
|
Lee YH, Stallcup MR. Minireview: protein arginine methylation of nonhistone proteins in transcriptional regulation. Mol Endocrinol 2009; 23:425-33. [PMID: 19164444 DOI: 10.1210/me.2008-0380] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Endocrine regulation frequently culminates in altered transcription of specific genes. The signal transduction pathways, which transmit the endocrine signal from cell surface to the transcription machinery, often involve posttranslational modifications of proteins. Although phosphorylation has been by far the most widely studied protein modification, recent studies have indicated important roles for other types of modification, including protein arginine methylation. Ten different protein arginine methyltransferase (PRMT) family members have been identified in mammalian cells, and numerous substrates are being identified for these PRMTs. Whereas major attention has been focused on the methylation of histones and its role in chromatin remodeling and transcriptional regulation, there are many nonhistone substrates methylated by PRMTs. This review primarily focuses on recent progress on the roles of the nonhistone protein methylation in transcription. Protein methylation of coactivators, transcription factors, and signal transducers, among other proteins, plays important roles in transcriptional regulation. Protein methylation may affect protein-protein interaction, protein-DNA or protein-RNA interaction, protein stability, subcellular localization, or enzymatic activity. Thus, protein arginine methylation is critical for regulation of transcription and potentially for various physiological/pathological processes.
Collapse
Affiliation(s)
- Young-Ho Lee
- Department of Biochemistry and Molecular Biology, Norris Comprehensive Cancer Center, Los Angeles, California 90089-9176, USA
| | | |
Collapse
|
304
|
Spreafico M, Ernst B, Lill M, Smiesko M, Vedani A. Mixed‐Model QSAR at the Glucocorticoid Receptor: Predicting the Binding Mode and Affinity of Psychotropic Drugs. ChemMedChem 2009; 4:100-9. [DOI: 10.1002/cmdc.200800274] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Morena Spreafico
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel (Switzerland), Fax: (+41) 61‐267‐1552
| | - Beat Ernst
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel (Switzerland), Fax: (+41) 61‐267‐1552
| | - Markus A. Lill
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907 (USA)
| | - Martin Smiesko
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel (Switzerland), Fax: (+41) 61‐267‐1552
| | - Angelo Vedani
- Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel (Switzerland), Fax: (+41) 61‐267‐1552
| |
Collapse
|
305
|
Wang Y, Zong H, Chi Y, Hong Y, Yang Y, Zou W, Yun X, Gu J. Repression of estrogen receptor alpha by CDK11p58 through promoting its ubiquitin-proteasome degradation. J Biochem 2009; 145:331-43. [PMID: 19122208 DOI: 10.1093/jb/mvn177] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Estrogen receptor alpha (ERalpha) is a ligand-dependent transcription factor that mediates physiological responses to 17beta-estradiol (E(2)). These responses of cells to estrogen are regulated in part by degradation of ERalpha. In this report, we found that CDK11(p58) repressed ERalpha transcriptional activity. And we further demonstrated that ERalpha protein level was down-regulated by CDK11(p58) in mammalian cells in a ligand independent manner. This effect could be abrogated by treatment with proteasome inhibitor MG132. Our results indicated that the ubiquitin/proteasome-mediated degradation of ERalpha was promoted by CDK11(p58). Furthermore, the interaction between ERalpha and CDK11(p58) was detected. This interaction was necessary for the polyubiquitination and degradation of ERalpha. On the contrary, the other isoform of CDK11, CDK11(p110) and the kinase dead mutant of CDK11(p58), D224N, did not associate with ERalpha and failed to reduce the ERalpha protein level. These data identified a new negative regulatory protein of ERalpha and provided a new pathway by which CDK11(p58) negatively regulated cells.
Collapse
Affiliation(s)
- Yanlin Wang
- Gene Research Center, Shanghai Medical College and Institutes of Biomedical, Shanghai 200032, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
306
|
Herr I, Büchler MW, Mattern J. Glucocorticoid-mediated apoptosis resistance of solid tumors. Results Probl Cell Differ 2009; 49:191-218. [PMID: 19132324 DOI: 10.1007/400_2008_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
More than a quarter of a century ago, the phenomenon of glucocorticoid-induced apoptosis in the majority of hematological cells was first recognized. More recently, glucocorticoid-induced antiapoptotic signaling associated with apoptosis resistance towards cytotoxic therapy has been identified in cells of epithelial origin, most of malignant solid tumors and some other tissues. Despite these huge amounts of data demonstrating differential pro- and anti-apoptotic effects of glucocorticoids, the underlying mechanisms of cell type-specific glucocorticoid signaling are just beginning to be described. This review summarizes our present understanding of cell type-specific pro- and anti-apoptotic signaling induced by glucocorticoids. We shortly introduce mechanisms of glucocorticoid resistance of hematological cells. We highlight and discuss the emerging molecular evidence of a general induction of survival signaling in epithelial cells and carcinoma cells by glucocorticoids. We give a summary of our current knowledge of decreased proliferation rates in response to glucocorticoid pre- and combination treatment, which are suspicious to be involved not only in protection of normal tissues, but also in protection of solid tumors from cytotoxic effects of anticancer agents.
Collapse
Affiliation(s)
- Ingrid Herr
- Department of Surgery, University of Heidelberg, Germany.
| | | | | |
Collapse
|
307
|
Abstract
Regulating gene expression is a complex process requiring the interaction of multiple transcription factors with their cognate recognition sequences. While these DNA-bound transcription factors are the primary drivers of gene expression, the capacity of a transcription factor to alter gene expression is tempered by its association with a host of coregulatory proteins that are recruited to the DNA-bound transcription factor. We have developed a novel approach to isolate large complexes of proteins associated with the DNA-bound estrogen receptor alpha (ERalpha) using an agarose-based electrophoretic mobility shift assay (EMSA). This method should be readily adapted to a variety of cultured cell lines, DNA sequences, and transcription factors and has the potential to provide valuable information about a wide variety of regulatory proteins involved in influencing gene expression.
Collapse
|
308
|
Velarde RA, Robinson GE, Fahrbach SE. Coordinated responses to developmental hormones in the Kenyon cells of the adult worker honey bee brain (Apis mellifera L.). JOURNAL OF INSECT PHYSIOLOGY 2009; 55:59-69. [PMID: 19013465 DOI: 10.1016/j.jinsphys.2008.10.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 10/14/2008] [Accepted: 10/15/2008] [Indexed: 05/27/2023]
Abstract
The brains of experienced forager honey bees exhibit predictable changes in structure, including significant growth of the neuropil of the mushroom bodies. In vertebrates, members of the superfamily of nuclear receptors function as key regulators of neuronal structure. The adult insect brain expresses many members of the nuclear receptor superfamily, suggesting that insect neurons are also likely important targets of developmental hormones. The actions of developmental hormones (the ecdysteroids and the juvenile hormones) in insects have been primarily explored in the contexts of metamorphosis and vitellogenesis. The cascade of gene expression activated by 20-hydroxyecdysone and modulated by juvenile hormone is strikingly conserved in these different physiological contexts. We used quantitative RT-PCR to measure, in the mushroom bodies of the adult worker honey bee brain, relative mRNA abundances of key members of the nuclear receptor superfamily (EcR, USP, E75, Ftz-f1, and Hr3) that participate in the metamorphosis/vitellogenesis cascade. We measured responses to endogenous peaks of hormones experienced early in adult life and to exogenous hormones. Our studies demonstrate that a population of adult insect neurons is responsive to endocrine signals through the use of conserved portions of the canonical ecdysteroid transcriptional cascade previously defined for metamorphosis and vitellogenesis.
Collapse
Affiliation(s)
- Rodrigo A Velarde
- Department of Biology, Wake Forest University, Box 7325, Winston-Salem, NC 27109, USA.
| | | | | |
Collapse
|
309
|
Ismail P, Abbasi S, Azimi C, Othman F, Noori Dalo M, Ashtiani Z, Mojarrad M, Oskouei S, Nejad F. Estrogen Receptor-α Gene Codon 10 (T392C) Polymorphism in Iranian Women with Breast Cancer: A Case Study. ACTA ACUST UNITED AC 2009. [DOI: 10.3923/tms.2009.1.10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
310
|
Fehrenbacher JC, Loverme J, Clarke W, Hargreaves KM, Piomelli D, Taylor BK. Rapid pain modulation with nuclear receptor ligands. ACTA ACUST UNITED AC 2008; 60:114-24. [PMID: 19162071 DOI: 10.1016/j.brainresrev.2008.12.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2008] [Indexed: 12/13/2022]
Abstract
We discuss and present new data regarding the physiological and molecular mechanisms of nuclear receptor activation in pain control, with a particular emphasis on non-genomic effects of ligands at peroxisome proliferator-activated receptor (PPAR), GPR30, and classical estrogen receptors. PPARalpha agonists rapidly reduce both acute and chronic pain in a number of pain assays. These effects precede transcriptional anti-inflammatory actions, and are mediated in part by IK(ca) and BK(ca) channels on DRG neurons. In contrast to the peripheral site of action of PPARalpha ligands, the dorsal horn supports the expression of PPARgamma. Intrathecal administration of PPARgamma ligands rapidly (< or =5 min) attenuated mechanical and thermal hypersensitivity associated with nerve injury in a dose-dependent manner that could be blocked with PPARgamma antagonists. By contrast, a PPARgamma antagonist itself rapidly increased the mechanical allodynia associated with nerve injury. These data suggest that ligand-dependent, non-genomic activation of spinal PPARgamma decreases behavioral signs of inflammatory and neuropathic pain. We also report that the GPR30 is expressed on cultured sensory neurons, that activation of the receptor elicits signaling to increase calcium accumulation. This signaling may contribute to increased neuronal sensitivity as treatment with the GPR30 agonist induces hyperalgesia. Finally, application of the membrane-impermeable 17beta-E(2)-BSA rapidly (within 15 min) enhanced BK-stimulated inositol phosphate (IP) accumulation and PGE(2)-mediated cAMP accumulation in trigeminal ganglion cultures. We conclude that nuclear receptor ligands may operate through rapid, non-genomic mechanisms to modulate inflammatory and neuropathic pain.
Collapse
Affiliation(s)
- Jill C Fehrenbacher
- Department of Endodontics, University of Texas Health Science Center, San Antonio, TX, USA
| | | | | | | | | | | |
Collapse
|
311
|
|
312
|
Kim JY, Son YL, Lee YC. Involvement of SMRT corepressor in transcriptional repression by the vitamin D receptor. Mol Endocrinol 2008; 23:251-64. [PMID: 19098224 DOI: 10.1210/me.2008-0426] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
To repress the expression of target genes, the unliganded nuclear receptor generally recruits the silencing mediator of retinoid and thyroid hormone receptor (SMRT)/nuclear receptor corepressor via its direct association with the conserved motif within bipartite nuclear receptor-interaction domains (IDs) of the corepressor. Here, we investigated the involvement of the SMRT corepressor in transcriptional repression by the unliganded vitamin D receptor (VDR). Using small interference RNA against SMRT in human embryonic kidney 293 cells, we demonstrated that SMRT is involved in the repression of the VDR-target genes, osteocalcin and vitamin D(3) 24-hydroxylase in vivo. Consistent with this, VDR and SMRT are recruited to the vitamin D response element of the endogenous osteocalcin promoter in the absence of 1alpha,25-(OH)(2)D(3) in chromatin immunoprecipitation assays. To address the involvement of the VDR-specific interaction of SMRT in this repression, we identified the molecular determinants of the interaction between VDR and SMRT. Interestingly, VDR specifically interacts with ID1 of the SMRT/nuclear receptor corepressor and that ID1 is required for their stable interaction. We also identified specific residues in the SMRT-ID1 that are required for VDR binding, using the one- plus two-hybrid system, a novel genetic selection method for specific missense mutations that disrupt protein-protein interactions. These mutational studies revealed that VDR interaction requires a wide range of the residues within and outside the extended helix motif of SMRT-ID1. Notably, SMRT mutants defective in the VDR interaction were also defective in the repression of endogenous VDR-target genes, indicating that the SMRT corepressor is directly involved in the VDR-mediated repression in vivo via an ID1-specific interaction with the VDR.
Collapse
Affiliation(s)
- Ji Young Kim
- School of Biological Sciences and Technology, Chonnam National University, Buk-gu, Gwangju 500-757, South Korea
| | | | | |
Collapse
|
313
|
Álvarez L, Martí M, Veleiro A, Misico R, Estrin D, Pecci A, Burton G. Hemisuccinate of 21-Hydroxy-6,19-Epoxyprogesterone: A Tissue-Specific Modulator of the Glucocorticoid Receptor. ChemMedChem 2008; 3:1869-77. [DOI: 10.1002/cmdc.200800256] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
314
|
Hossain A, Hajman K, Charitidi K, Erhardt S, Zimmermann U, Knipper M, Canlon B. Prenatal dexamethasone impairs behavior and the activation of the BDNF exon IV promoter in the paraventricular nucleus in adult offspring. Endocrinology 2008; 149:6356-65. [PMID: 18755799 DOI: 10.1210/en.2008-0388] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prenatal manipulations to the hypothalamic-pituitary-adrenal axis are shown to affect auditory responses to an acoustic challenge as well as behavior in adult life. To achieve these results, we examined the effect of prenatal dexamethasone (DEX) treatment in male and female adult rat offspring by assessing body and adrenal weight, anxiety using the elevated plus maze (EPM), and acoustic startle responses as well as the effects of acoustic challenge in the paraventricular nucleus (PVN). DEX male offspring had reduced adrenal gland weight in adult life and demonstrated anxiolytic-like behavior when tested on the EPM. The acoustic startle amplitude in naive DEX-treated male offspring was significantly higher compared with saline (SAL)-treated males and females and DEX-treated females. When challenged with either a glucocorticoid agonist or antagonist, the startle response of the SAL-treated males and females significantly increased or decreased in the presence of agonist and antagonist treatment, respectively, whereas DEX males and females were not affected. Acoustic challenge caused an increase in c-fos mRNA and glucocorticoid receptor nuclear translocation in the PVN of all groups. BDNF and TrkB mRNA increased in the PVN after acoustic challenge in the SAL-treated males and females but not in the DEX males or females. These findings exemplify the differential sensitivity of the developing nervous and endocrine systems to prenatal hormonal stress and demonstrate that prenatal DEX treatment elicits long-term behavioral alterations related to anxiety and auditory processing.
Collapse
Affiliation(s)
- Amzad Hossain
- Department of Physiology & Pharmacology Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
315
|
Zhang J, Ge R, Matte-Martone C, Goodwin J, Shlomchik WD, Mamula MJ, Kooshkabadi A, Hardy MP, Geller D. Characterization of a novel gain of function glucocorticoid receptor knock-in mouse. J Biol Chem 2008; 284:6249-59. [PMID: 19017639 DOI: 10.1074/jbc.m807997200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucocorticoids (GCs) exert profound influences on many physiologic functions by virtue of their diverse roles in growth, development, and maintenance of homeostasis. We previously created a novel gain of function in the human glucocorticoid receptor (hGR), hGRM604L, which is active at GC concentrations 5-10-fold lower than wild-type GR. To gain a greater insight into GC physiology in vivo, we inserted this mutant GR (GRM610L in mice) into mice via homologous recombination. Mice expressing the allele are phenotypically normal with respect to GC function. However, corticosterone levels, ACTH levels, and adrenocortical size are markedly reduced, suggesting they are phenotypically normal because the mutant GR alters the basal regulation of the hypothalamic-pituitary-adrenal axis. We demonstrate via physiologic and immunologic studies that GRM610L mice have increased sensitivity to GCs in vivo. Sensitivity to the actions of endogenous GCs may be an important factor underlying the development of many human diseases including hypertension, obesity, and diabetes. Our model may provide a new and powerful tool for the study of GC physiological and pathological processes in vivo.
Collapse
Affiliation(s)
- Junhui Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520-8029, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
316
|
Kruse SW, Suino-Powell K, Zhou XE, Kretschman JE, Reynolds R, Vonrhein C, Xu Y, Wang L, Tsai SY, Tsai MJ, Xu HE. Identification of COUP-TFII orphan nuclear receptor as a retinoic acid-activated receptor. PLoS Biol 2008; 6:e227. [PMID: 18798693 PMCID: PMC2535662 DOI: 10.1371/journal.pbio.0060227] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Accepted: 08/04/2008] [Indexed: 12/22/2022] Open
Abstract
The chicken ovalbumin upstream promoter-transcription factors (COUP-TFI and II) make up the most conserved subfamily of nuclear receptors that play key roles in angiogenesis, neuronal development, organogenesis, cell fate determination, and metabolic homeostasis. Although the biological functions of COUP-TFs have been studied extensively, little is known of their structural features or aspects of ligand regulation. Here we report the ligand-free 1.48 Å crystal structure of the human COUP-TFII ligand-binding domain. The structure reveals an autorepressed conformation of the receptor, where helix α10 is bent into the ligand-binding pocket and the activation function-2 helix is folded into the cofactor binding site, thus preventing the recruitment of coactivators. In contrast, in multiple cell lines, COUP-TFII exhibits constitutive transcriptional activity, which can be further potentiated by nuclear receptor coactivators. Mutations designed to disrupt cofactor binding, dimerization, and ligand binding, substantially reduce the COUP-TFII transcriptional activity. Importantly, retinoid acids are able to promote COUP-TFII to recruit coactivators and activate a COUP-TF reporter construct. Although the concentration needed is higher than the physiological levels of retinoic acids, these findings demonstrate that COUP-TFII is a ligand-regulated nuclear receptor, in which ligands activate the receptor by releasing it from the autorepressed conformation. Unlike other classes of receptors, nuclear receptors can bind directly to DNA and act as transcription factors, playing key roles in embryonic development and cellular metabolism. Most nuclear receptors are activated by signal-triggering molecules (ligands) and can regulate their activity by recruiting coactivator proteins. However, the ligands are unknown for a subset of “orphan” nuclear receptors, including the chicken ovalbumin promoter-transcription factors (COUP-TFI and II, and EAR2). COUP-TFs are the most conserved nuclear receptors, with roles in angiogenesis, neuronal development, organogenesis, and metabolic homeostasis. Here we demonstrate that COUP-TFII is a ligand-regulated nuclear receptor that can be activated by unphysiological micromolar concentrations of retinoic acids. We determined the structure of the ligand-free ligand-binding domain of the human COUP-TFII, revealing the autorepressed conformation of the receptor, where helix α10 is bent into the ligand-binding pocket and the activation function-2 helix is folded into the cofactor binding site, thus preventing the recruitment of coactivators. These results suggest a mechanism where ligands activate COUP-TFII by releasing the receptor from the autorepressed conformation. The identification of COUP-TFII as a low-affinity retinoic acid receptor suggests ways of searching for the endogenous ligands that may ultimately link retinoic acid and COUP-TF signaling pathways. Structural and functional studies reveal that the orphan nuclear receptor COUP-TFII is a low-affinity receptor for retinoic acids. paving the way to finding the endogenous ligands that may ultimately link retinoic acid and COUP-TF signaling pathways.
Collapse
Affiliation(s)
- Schoen W Kruse
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Kelly Suino-Powell
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - X. Edward Zhou
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Jennifer E Kretschman
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Ross Reynolds
- Department of Physics, Grand Valley State University, Allendale, Michigan, United States of America
| | - Clemens Vonrhein
- Global Phasing Ltd., Sheraton House, Castle Park, Cambridge, United Kingdom
| | - Yong Xu
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Liliang Wang
- Department of Molecular and Cellular Biology and Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sophia Y Tsai
- Department of Molecular and Cellular Biology and Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ming-Jer Tsai
- Department of Molecular and Cellular Biology and Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, United States of America
| | - H. Eric Xu
- Laboratory of Structural Sciences, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
317
|
Rantham Prabhakara JP, Feist G, Thomasson S, Thompson A, Schommer E, Ghribi O. Differential effects of 24-hydroxycholesterol and 27-hydroxycholesterol on tyrosine hydroxylase and alpha-synuclein in human neuroblastoma SH-SY5Y cells. J Neurochem 2008; 107:1722-9. [PMID: 19014385 PMCID: PMC3205449 DOI: 10.1111/j.1471-4159.2008.05736.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Evidence suggests that environmental and dietary factors may contribute to the pathogenesis of Parkinson's disease (PD). High dietary intake of cholesterol is such a factor that has been shown to increase or decrease the risk of PD. However, because circulating cholesterol does not cross the blood-brain barrier, the mechanisms linking dietary cholesterol to the pathogenesis of PD remain to be understood. In contrast to cholesterol, the oxidized cholesterol metabolites (oxysterols), 24S-hydroxycholesterol (24-OHC) and 27-hydroxycholesterol (27-OHC), can cross the blood-brain barrier and may place the brain at risk of degeneration. In this study, we incubated the human neuroblastoma SH-SY5Y cells for 24 h with 24-OHC, 27-OHC, or a mixture of 24-OHC plus 27-OHC, and have determined effects on tyrosine hydroxylase (the rate-limiting enzyme in dopamine synthesis) levels, alpha-synuclein levels, and apoptosis. We demonstrate that while 24-OHC increases the levels of tyrosine hydroxylase, 27-OHC increases levels of alpha-synuclein, and induces apoptosis. Our findings show for the first time that oxysterols trigger changes in levels of proteins that are associated with the pathogenesis of PD. As steady state levels of 24-OHC and 27-OHC are tightly regulated in the brain, disturbances in these levels may contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Jaya Prasanthi Rantham Prabhakara
- Department of Pharmacology, Physiology and Therapeutics, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58202, USA
| | | | | | | | | | | |
Collapse
|
318
|
Esposito T, Dominguez P, Varriale B. Hormonal regulation and characterisation of the aldehyde oxidase-like gene of hamster Harderian gland. J Steroid Biochem Mol Biol 2008; 112:157-63. [PMID: 18848890 DOI: 10.1016/j.jsbmb.2008.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 07/01/2008] [Accepted: 09/16/2008] [Indexed: 10/21/2022]
Abstract
The HG is a compound tubulo-alveolar gland located in the orbital cavity of the majority of vertebrates. In the golden hamster it shows a clear cut sexual dimorphism in both morphological and biochemical parameters such as cell types, protein pattern, lipid metabolism, porphyrin content, steroid hormone receptor expression. In a previous study we found that in primary culture of male hamster Harderian gland (HG), androgens (A) increase the MHG07 (male Harderian gland) expression and this effect is abrogated by both flutamide and cycloheximide. The present study represents a deeper analysis on MHG07 regulation by other members of steroid/thyroid hormone superfamily. Estrogens (E) impair the stimulatory effect of A and after the addition of a pure anti-estrogen, ICI 164,384, the negative effect of E is abrogated. Dexamethasone (Dex), used alone or in combination with A negatively affect the MHG07 expression. Also T(3) increases the expression of MHG07 mRNA. Progesterone (P) does not affect the expression of MHG07 mRNA. The use of cycloheximide abrogates the effect of steroids, suggesting that the latter act through their own receptors. Dose-response experiments show that low steroid concentrations (10(-12)M) are sufficient to affect the MHG07 expression. It is argued that the expression of MHG07 is under a highly coordinate relationship between androgen, estrogen, glucocorticoid, retinoic acid and thyroid hormones.
Collapse
Affiliation(s)
- Teresa Esposito
- Department of Experimental Medicine, Faculty of Medicine, II University of Naples, Naples, Italy
| | | | | |
Collapse
|
319
|
Yang X, Song L, Xue HW. Membrane steroid binding protein 1 (MSBP1) stimulates tropism by regulating vesicle trafficking and auxin redistribution. MOLECULAR PLANT 2008; 1:1077-87. [PMID: 19825605 DOI: 10.1093/mp/ssn071] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Overexpression of membrane steroid binding protein 1 (MSBP1) stimulates the root gravitropism and anti-gravitropism of hypocotyl, which is mainly due to the enhanced auxin redistribution in the bending regions of hypocotyls and root tips. The inhibitory effects by 1-N-naphthylphthalamic acid (NPA), an inhibitor of polar auxin transport, are suppressed under the MSBP1 overexpression, suggesting the positive effects of MSBP1 on polar auxin transport. Interestingly, sub-cellular localization studies showed that MSBP1 is also localized in endosomes and observations of the membrane-selective dye FM4-64 revealed the enhanced vesicle trafficking under MSBP1 overexpression. MSBP1-overexpressing seedlings are less sensitive to brefeldin A (BFA) treatment, whereas the vesicle trafficking was evidently reduced by suppressed MSBP1 expression. Enhanced MSBP1 does not affect the polar localization of PIN2, but stimulates the PIN2 cycling and enhances the asymmetric PIN2 redistribution under gravi-stimulation. These results suggest that MSBP1 could enhance the cycling of PIN2-containing vesicles to stimulate the auxin redistribution under gravi-stimulation, providing informative hints on interactions between auxin and steroid binding protein.
Collapse
Affiliation(s)
- Xi Yang
- Chinese Academy of Sciences, 200032 Shanghai, China
| | | | | |
Collapse
|
320
|
McKeen HD, McAlpine K, Valentine A, Quinn DJ, McClelland K, Byrne C, O'Rourke M, Young S, Scott CJ, McCarthy HO, Hirst DG, Robson T. A novel FK506-like binding protein interacts with the glucocorticoid receptor and regulates steroid receptor signaling. Endocrinology 2008; 149:5724-34. [PMID: 18669603 DOI: 10.1210/en.2008-0168] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
FKBP-like (FKBPL) protein is a novel immunophilin-like protein that plays a role in the cellular stress response. Its three tetratricopeptide repeat motifs are homologous to the heat shock protein 90 interaction sites of other immunophilins that have roles in steroid hormone receptor signaling. In this study, using biomolecular complementation and coimmunoprecipitation techniques, we show that FKBPL also colocalizes and interacts with the components of the heat shock protein 90-glucocorticoid receptor (GR) complex and demonstrate that the PPIase domain of FKBPL is important for the interaction between this complex and the dynein motor protein, dynamitin. Treatment of DU145 cells with the GR ligand, dexamethasone, induced a rapid and coordinated translocation of both GR and FKBPL to the nucleus; this response was perturbed when FKBPL was knocked down with a targeted small interfering RNA. Furthermore, overexpression of FKBPL increased GR protein levels and transactivation of a luciferase reporter gene in response to dexamethasone in DU145 cells. However, these responses were cell line dependent. In summary, these data suggest that FKBPL can be classed as a new member of the FKBP protein family with a role in steroid receptor complexes and signaling.
Collapse
Affiliation(s)
- Hayley D McKeen
- School of Pharmacy, Queen's University, Belfast BT9 7BL, Northern Ireland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
321
|
Schultz-Norton JR, Ziegler YS, Likhite VS, Yates JR, Nardulli AM. Isolation of novel coregulatory protein networks associated with DNA-bound estrogen receptor alpha. BMC Mol Biol 2008; 9:97. [PMID: 18973695 PMCID: PMC2585101 DOI: 10.1186/1471-2199-9-97] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 10/30/2008] [Indexed: 12/19/2022] Open
Abstract
Background DNA-bound transcription factors recruit an array of coregulatory proteins that influence gene expression. We previously demonstrated that DNA functions as an allosteric modulator of estrogen receptor α (ERα) conformation, alters the recruitment of regulatory proteins, and influences estrogen-responsive gene expression and reasoned that it would be useful to develop a method of isolating proteins associated with the DNA-bound ERα using full-length receptor and endogenously-expressed nuclear proteins. Results We have developed a novel approach to isolate large complexes of proteins associated with the DNA-bound ERα. Purified ERα and HeLa nuclear extracts were combined with oligos containing ERα binding sites and fractionated on agarose gels. The protein-DNA complexes were isolated and mass spectrometry analysis was used to identify proteins associated with the DNA-bound receptor. Rather than simply identifying individual proteins that interact with ERα, we identified interconnected networks of proteins with a variety of enzymatic and catalytic activities that interact not only with ERα, but also with each other. Characterization of a number of these proteins has demonstrated that, in addition to their previously identified functions, they also influence ERα activity and expression of estrogen-responsive genes. Conclusion The agarose gel fractionation method we have developed would be useful in identifying proteins that interact with DNA-bound transcription factors and should be easily adapted for use with a variety of cultured cell lines, DNA sequences, and transcription factors.
Collapse
Affiliation(s)
- Jennifer R Schultz-Norton
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | | | | | | | | |
Collapse
|
322
|
Mo B, Callegari E, Telefont M, Renner KJ. Estrogen regulation of proteins in the rat ventromedial nucleus of the hypothalamus. J Proteome Res 2008; 7:5040-8. [PMID: 18841879 DOI: 10.1021/pr8005974] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The effects of estradiol (E2) on the expression of proteins in the pars lateralis of the ventromedial nucleus of the hypothalamus (VMNpl) in ovariectomized rats was studied using 2-dimensional gel electrophoresis followed by RPLC-nanoESI-MS/MS. E2 treatment resulted in the up-regulation of 29 identified proteins. Many of these proteins are implicated in the promotion of neuronal plasticity and signaling.
Collapse
Affiliation(s)
- Bing Mo
- Department of Biology and Neuroscience Group, University of South Dakota, Vermillion, South Daklota 57069, USA
| | | | | | | |
Collapse
|
323
|
Kamiyama K, Matsuda N, Yamamoto S, Takano KI, Takano Y, Yamazaki H, Kageyama SI, Yokoo H, Nagata T, Hatakeyama N, Tsukada K, Hattori Y. Modulation of glucocorticoid receptor expression, inflammation, and cell apoptosis in septic guinea pig lungs using methylprednisolone. Am J Physiol Lung Cell Mol Physiol 2008; 295:L998-L1006. [PMID: 18836031 DOI: 10.1152/ajplung.00459.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The use of glucocorticoids for treatment of sepsis has waxed and waned during the past several decades, and recent randomized controlled trials have evoked a reassessment of this therapy. Most glucocorticoid actions are mediated by its specific intracellular receptors (GRs). Thus we initially evaluated whether sepsis and high-dose corticosteroid therapy can regulate guinea pig pulmonary expression of GRs: active receptor, GRalpha, and dominant negative receptor, GRbeta. Sepsis induction by LPS injection (300 mug/kg ip) decreased mRNA and protein levels of GRalpha and increased protein expression of GRbeta in lungs. High-dose methylprednisolone (40 mg/kg ip), administered simultaneously with LPS, markedly potentiated the decrease in GRalpha expression but slightly affected the increase in GRbeta expression. Consequently, this led to a significant reduction in GRalpha nuclear translocation. Nevertheless, methylprednisolone treatment strongly eliminated LPS induction of NF-kappaB activity, as determined by NF-kappaB nuclear translocation and by gel mobility shift assays. Furthermore, the LPS-induced increase in inflammatory cells in bronchoalveolar lavage fluid was blunted by administration of the corticosteroid. On the other hand, immunofluorescent staining for cleaved caspase-3 showed a marked increase in this proapoptotic marker in lung sections, and terminal deoxynucleotidyl transferase dUTP-mediated nick-end labeling (TUNEL) represented an enhanced appearance of cell apoptosis in lungs and spleen when methylprednisolone was given together with LPS. Cell apoptosis is now considered to play a role in the pathogenesis of septic syndrome. We thus suggest that the action of glucocorticoids at high doses to accelerate sepsis-induced cell apoptosis may overwhelm their therapeutic advantages in septic shock.
Collapse
Affiliation(s)
- Koki Kamiyama
- Dept. of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences, Univ. of Toyama, Toyama 930-0194, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
324
|
Wong HY, Hoogerbrugge JW, Pang KL, van Leeuwen M, van Royen ME, Molier M, Berrevoets CA, Dooijes D, Dubbink HJ, van de Wijngaart DJ, Wolffenbuttel KP, Trapman J, Kleijer WJ, Drop SLS, Grootegoed JA, Brinkmann AO. A novel mutation F826L in the human androgen receptor in partial androgen insensitivity syndrome; increased NH2-/COOH-terminal domain interaction and TIF2 co-activation. Mol Cell Endocrinol 2008; 292:69-78. [PMID: 18656523 DOI: 10.1016/j.mce.2008.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2008] [Revised: 06/26/2008] [Accepted: 06/27/2008] [Indexed: 10/21/2022]
Abstract
A novel mutation F826L located within the ligand binding domain (LBD) of the human androgen receptor (AR) was investigated. This mutation was found in a boy with severe penoscrotal hypospadias (classified as 46,XY DSD). The AR mutant F826L appeared to be indistinguishable from the wild-type AR, with respect to ligand binding affinity, transcriptional activation of MMTV-luciferase and ARE2-TATA-luciferase reporter genes, protein level in genital skin fibroblasts (GSFs), and sub-cellular distribution in transfected cells. However, an at least two-fold higher NH2-/COOH-terminal domain interaction was found in luciferase and GST pull-down assays. A two-fold increase was also observed for TIF2 (transcription intermediary factor 2) co-activation of the AR F826L COOH-terminal domain. This increase could not be explained by a higher stability of the mutant protein, which was within wild-type range. Repression of transactivation by the nuclear receptor co-repressor (N-CoR) was not affected by the AR F826L mutation. The observed properties of AR F826L would be in agreement with an increased activity rather than with a partial defective AR transcriptional activation. It is concluded that the penoscrotal hypospadias in the present case is caused by an as yet unknown mechanism, which still may involve the mutant AR.
Collapse
Affiliation(s)
- Hao Yun Wong
- Department of Reproduction and Development, Erasmus MC, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Stahl S, Davies MR, Cook DI, Graham MJ. Nuclear hormone receptor-dependent regulation of hepatic transporters and their role in the adaptive response in cholestasis. Xenobiotica 2008; 38:725-77. [DOI: 10.1080/00498250802105593] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
326
|
Shimizu N, Yoshikawa N, Wada T, Handa H, Sano M, Fukuda K, Suematsu M, Sawai T, Morimoto C, Tanaka H. Tissue- and context-dependent modulation of hormonal sensitivity of glucocorticoid-responsive genes by hexamethylene bisacetamide-inducible protein 1. Mol Endocrinol 2008; 22:2609-23. [PMID: 18801933 DOI: 10.1210/me.2008-0101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Physiological and pharmacological processes mediated by glucocorticoids involve tissue- and context-specific regulation of glucocorticoid-responsive gene expression via glucocorticoid receptor (GR). However, the molecular mechanisms underlying such highly coordinated regulation of glucocorticoid actions remain to be studied. We here addressed this issue using atp1a1 and scnn1a, both of which are up-regulated in response to corticosteroids in human embryonic kidney-derived 293 cells, but resistant in liver-derived HepG2 cells. Hexamethylene bisacetamide-inducible protein 1 (HEXIM1) represses gene expression via, at least, two distinct mechanisms, i.e. positive transcription elongation factor b sequestration and direct interaction with GR, and is relatively high in HepG2 cells compared with 293 cells. Given this, we focused on the role of HEXIM1 in transcriptional regulation of these GR target genes. In HepG2 cells, hormone resistance of atp1a1 and scnn1a was diminished by either knockdown of HEXIM1 or overexpression of GR. Such a positive effect of exogenous expression of GR was counteracted by concomitant overexpression of HEXIM1, indicating the balance between GR and HEXIM1 modulates hormonal sensitivity of these genes. In support of this, the hormone-dependent recruitment of RNA polymerase II onto atp1a1 promoter was in parallel with that of GR. Moreover, we revealed that not positive transcription elongation factor b-suppressing activity but direct interaction with GR of HEXIM1 plays a major role in suppression of promoter recruitment of the receptor and subsequent atp1a1 and scnn1a gene activation. Collectively, we may conclude that HEXIM1 may participate in tissue-selective determination of glucocorticoid sensitivity via direct interaction with GR at least in certain gene sets including atp1a1 and scnn1a.
Collapse
Affiliation(s)
- Noriaki Shimizu
- Division of Clinical Immunology, Advanced Clinical Research Center, Research Hospital, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
327
|
Wood J, Lee SS, Hague A. Bag-1 proteins in oral squamous cell carcinoma. Oral Oncol 2008; 45:94-102. [PMID: 18804403 DOI: 10.1016/j.oraloncology.2008.07.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 07/17/2008] [Accepted: 07/17/2008] [Indexed: 10/21/2022]
Abstract
Bag-1 is an anti-apoptotic protein that exhibits altered expression in many malignancies, including oral squamous cell carcinoma. The bag-1 gene gives rise to different protein products with different subcellular localisations through alternative translational initiation sites. In oral squamous cell carcinoma, cytoplasmic expression has been associated with metastasis to regional lymph nodes and poor prognosis. In contrast, the longest Bag-1 isoform is nuclear and may regulate differentiation in oral epithelium. In this review, the functions of the three isoforms of Bag-1 expressed in oral epithelial cells are discussed in relation to their contribution to oral carcinogenesis.
Collapse
Affiliation(s)
- Jemma Wood
- Department of Oral and Dental Science, University of Bristol, Lower Maudlin Street, Bristol, BS1 2LY, UK
| | | | | |
Collapse
|
328
|
Carroll SM, Bridgham JT, Thornton JW. Evolution of hormone signaling in elasmobranchs by exploitation of promiscuous receptors. Mol Biol Evol 2008; 25:2643-52. [PMID: 18799714 DOI: 10.1093/molbev/msn204] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Specific interactions among proteins, nucleic acids, and metabolites drive virtually all cellular functions and underlie phenotypic complexity and diversity. Despite the fundamental importance of interactions, the mechanisms and dynamics by which they evolve are poorly understood. Here we describe novel interactions between a lineage-specific hormone and its receptors in elasmobranchs, a subclass of cartilaginous fishes, and infer how these associations evolved using phylogenetic and protein structural analyses. The hormone 1alpha-hydroxycorticosterone (1alpha-B) is a physiologically important steroid synthesized only in elasmobranchs. We show that 1alpha-B modulates gene expression in vitro by activating two paralogous intracellular transcription factors, the mineralocorticoid receptor (MR) and glucocorticoid receptor (GR), in the little skate Leucoraja erinacea; MR serves as a high-sensitivity and GR as a low-sensitivity receptor. Using functional analysis of extant and resurrected ancestral proteins, we show that receptor sensitivity to 1alpha-B evolved millions of years before the hormone itself evolved. The 1alpha-B differs from more ancient corticosteroids only by the addition of a hydroxyl group; the three-dimensional structure of the ancestral receptor shows that the ligand pocket contained ample unoccupied space to accommodate this moiety. Our findings indicate that the interactions between 1alpha-B and elasmobranch GR and MR proteins evolved by molecular exploitation: a novel hormone recruited into new functional partnerships two ancient receptors that had previously interacted with other ligands. The ancestral receptor's promiscuous capacity to fortuitously bind compounds that are slight structural variants of its original ligands set the stage for the evolution of this new interaction.
Collapse
|
329
|
Ali A, Balkovec JM, Greenlee M, Hammond ML, Rouen G, Taylor G, Einstein M, Ge L, Harris G, Kelly TM, Mazur P, Pandit S, Santoro J, Sitlani A, Wang C, Williamson J, Forrest MJ, Carballo-Jane E, Luell S, Lowitz K, Visco D. Discovery of betamethasone 17alpha-carbamates as dissociated glucocorticoid receptor modulators in the rat. Bioorg Med Chem 2008; 16:7535-42. [PMID: 18691892 DOI: 10.1016/j.bmc.2008.07.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/03/2008] [Accepted: 07/16/2008] [Indexed: 10/21/2022]
Abstract
A series of betamethasone 17alpha-carbamates were designed, synthesized, and evaluated for their ability to dissociate the two main functions of the glucocorticoid receptor, that is, transactivation and transrepression, in rat cell lines. A number of alkyl substituted betamethasone 17alpha-carbamates were identified with excellent affinity for the glucocorticoid receptor (e.g., 7, GR IC(50) 5.1 nM) and indicated dissociated profiles in functional assays of transactivation (rat tyrosine aminotransferase, TAT, and rat glutamine synthetase, GS) and transrepression (human A549 cells, MMP-1 assay). Gratifyingly, the in-vivo profile of these compounds, for example, 7, also indicated potent anti-inflammatory activity with impaired effects on glucose, insulin, triglycerides, and body weight. Taken together, these results indicate that dissociated glucocorticoid receptor modulators can be identified in rodents.
Collapse
Affiliation(s)
- Amjad Ali
- Department of Medicinal Chemistry, Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
330
|
Centenera MM, Harris JM, Tilley WD, Butler LM. The contribution of different androgen receptor domains to receptor dimerization and signaling. Mol Endocrinol 2008; 22:2373-82. [PMID: 18617596 DOI: 10.1210/me.2008-0017] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The androgen receptor (AR) is a ligand-activated transcription factor of the nuclear receptor superfamily that plays a critical role in male physiology and pathology. Activated by binding of the native androgens testosterone and 5alpha-dihydrotestosterone, the AR regulates transcription of genes involved in the development and maintenance of male phenotype and male reproductive function as well as other tissues such as bone and muscle. Deregulation of AR signaling can cause a diverse range of clinical conditions, including the X-linked androgen insensitivity syndrome, a form of motor neuron disease known as Kennedy's disease, and male infertility. In addition, there is now compelling evidence that the AR is involved in all stages of prostate tumorigenesis including initiation, progression, and treatment resistance. To better understand the role of AR signaling in the pathogenesis of these conditions, it is important to have a comprehensive understanding of the key determinants of AR structure and function. Binding of androgens to the AR induces receptor dimerization, facilitating DNA binding and the recruitment of cofactors and transcriptional machinery to regulate expression of target genes. Various models of dimerization have been described for the AR, the most well characterized interaction being DNA-binding domain- mediated dimerization, which is essential for the AR to bind DNA and regulate transcription. Additional AR interactions with potential to contribute to receptor dimerization include the intermolecular interaction between the AR amino terminal domain and ligand-binding domain known as the N-terminal/C-terminal interaction, and ligand-binding domain dimerization. In this review, we discuss each form of dimerization utilized by the AR to achieve transcriptional competence and highlight that dimerization through multiple domains is necessary for optimal AR signaling.
Collapse
Affiliation(s)
- Margaret M Centenera
- Dame Roma Mitchell Cancer Research Laboratories, Discipline of Medicine, The University of Adelaide and Hanson Institute, PO Box 14, Rundle Mall, Adelaide, South Australia 5000, Australia
| | | | | | | |
Collapse
|
331
|
Grasselli A, Nanni S, Colussi C, Aiello A, Benvenuti V, Ragone G, Moretti F, Sacchi A, Bacchetti S, Gaetano C, Capogrossi MC, Pontecorvi A, Farsetti A. Estrogen Receptor-α and Endothelial Nitric Oxide Synthase Nuclear Complex Regulates Transcription of Human Telomerase. Circ Res 2008; 103:34-42. [DOI: 10.1161/circresaha.107.169037] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We report that in endothelial cells, the angiogenic effect of 17β-estradiol (E2) is inhibited by the estrogen receptor (ER) antagonist ICI or the NO synthase (NOS) inhibitor 7-nitroindazole via downregulation of hTERT, the telomerase catalytic subunit, suggesting that E2 and NO are involved in controlling hTERT transcription. Quantitative Real-Time PCR and chromatin immunoprecipitations in E2-treated human umbilical vein endothelial cells, showed recruitment of ERs on the hTERT promoter and concomitant enrichment in histone 3 methylation at Lysine 79, a modification associated with transcription-competent chromatin. Confocal microscopy and re-chromatin immunoprecipitations revealed that on E2 induction, endothelial (e)NOS rapidly localized into the nucleus and associated with ERα on the hTERT promoter. Transfections of a constitutively active eNOS mutant (S1177D) strongly induced the hTERT promoter, indicating a direct role of the protein in hTERT transcriptional regulation. Mutation of the estrogen response element in the promoter abolished response to both ERs and active eNOS, demonstrating that the estrogen response element integrity is required for hTERT regulation by these factors. To investigate this novel regulation in a reduced NO environment, pulmonary endothelial cells were isolated from eNOS
−/−
mice and grown with/without E2. In wild-type cells, E2 significantly increased telomerase activity. In eNOS
−/−
cells, basal telomerase activity was rescued by exogenous eNOS or an NO donor, whereas responsiveness to E2 demanded the active protein. In conclusion, we document the novel findings of a combinatorial eNOS/ERα complex at the hTERT estrogen response element site and that active eNOS and ligand-activated ERs cooperate in regulating hTERT expression in the endothelium.
Collapse
Affiliation(s)
- Annalisa Grasselli
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Simona Nanni
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Claudia Colussi
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Aurora Aiello
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Valentina Benvenuti
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Gianluca Ragone
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Fabiola Moretti
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Ada Sacchi
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Silvia Bacchetti
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Carlo Gaetano
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Maurizio C. Capogrossi
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Alfredo Pontecorvi
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| | - Antonella Farsetti
- From the Department of Experimental Oncology (A.G., S.N., A.A., V.B., F.M., A.S., S.B., A.F.), Regina Elena Cancer Institute, Rome; Endocrinology (A.G., V.B., A.P.), Catholic University, Rome; Laboratory of Vascular Biology and Gene Therapy (S.N., C.C.), Centro Cardiologico “I. Monzino,” Milan; Laboratorio di Oncogenesi Molecolare (G.R.) and Laboratorio di Patologia Vascolare (C.G., M.C.C.), Istituto Dermopatico dell' Immacolata, Rome; and Institute of Neurobiology and Molecular Medicine (A.A., F.M
| |
Collapse
|
332
|
Li G, Lee YF, Liu S, Cai Y, Xie S, Liu NC, Bao BY, Chen Z, Chang C. Oxidative stress stimulates testicular orphan receptor 4 through forkhead transcription factor forkhead box O3a. Endocrinology 2008; 149:3490-9. [PMID: 18388194 PMCID: PMC2453090 DOI: 10.1210/en.2008-0121] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Early studies reveal that testicular orphan nuclear receptor 4 (TR4) modulates signaling pathways that control various cell functions. However, how TR4 activity is regulated without the involvement of specific ligand(s) remains unclear. Here we identify a daf-16 family protein-binding element (DBE; 5'-TGTTTAC-3') in the TR4 promoter that can be recognized by the forkhead transcriptional factor FOXO3a, a key stress-responsive factor, through which TR4 gene expression is activated. The interaction between DBE and FOXO3a was confirmed using EMSA and chromatin immunoprecipitation assays. Activation of FOXO3a by oxidative stress and phosphatidylinositol 3-kinase inhibitor induced TR4 expression; in contrast, suppression of FOXO3a by small interfering RNA can reduce oxidative stress-induced TR4 expression. The biological consequence of the FOXO3a-induced TR4 by oxidative stress is to protect against stress-induced cell death in which cells with reduced FOXO3a are less resistant to oxidative stress, and addition of functional TR4 can increase stress resistance. These results suggest that this new identified oxidative stress-FOXO3a-TR4 pathway is a fundamentally important mechanism regulating stress resistance and cell survival.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cell Line
- Cell Line, Tumor
- Cell Survival/drug effects
- Cells, Cultured
- Chromones/pharmacology
- Electrophoretic Mobility Shift Assay
- Forkhead Box Protein O3
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/physiology
- Gene Expression/drug effects
- Humans
- Hydrogen Peroxide/pharmacology
- Luciferases/genetics
- Luciferases/metabolism
- Male
- Mice
- Mice, Knockout
- Microscopy, Fluorescence
- Morpholines/pharmacology
- Oxidants/pharmacology
- Oxidative Stress
- Phosphorylation/drug effects
- Promoter Regions, Genetic/genetics
- Protein Binding
- RNA Interference
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- Testis/metabolism
Collapse
Affiliation(s)
- Gonghui Li
- George H. Whipple Laboratory for Cancer Research, Department of Pathology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
333
|
Hong J, Samudio I, Chintharlapalli S, Safe S. 1,1-bis(3'-indolyl)-1-(p-substituted phenyl)methanes decrease mitochondrial membrane potential and induce apoptosis in endometrial and other cancer cell lines. Mol Carcinog 2008; 47:492-507. [PMID: 18085536 PMCID: PMC2711558 DOI: 10.1002/mc.20407] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
1,1-Bis(3'-indolyl)-1-(p-substituted phenyl)methanes, containing p-t-butyl (DIM-C-pPhtBu) and phenyl (DIM-C-pPhC(6)H(5)) substituents, are peroxisome proliferator-activated receptor gamma (PPARgamma) agonists; however, DIM-C-pPhtBu-induced growth inhibition and cell death in human HEC1A endometrial cancer cells is PPARgamma-independent. DIM-C-pPhtBu decreased mitochondrial membrane potential (MMP) and promoted the release of cytochrome c and caspase activation and nuclear uptake of endonuclease G leading to apoptosis of HEC1A cells. DIM-C-pPhtBu specifically targeted the mitochondrial permeability transition pore complex (PTPC) because the DIM-C-pPhtBu-induced pro-apoptotic responses were inhibited by atractyloside (Atra), a compound that specifically interacts with the inner mitochondrial membrane adenine nucleotide transport (ANT) proteins. At the dose of Atra used in this study (300 microM), this compound alone did not alter the PTPC but inhibited the mitochondriotoxic effects of DIM-C-pPhtBu. DIM-C-pPhtBu/DIM-C-pPhC(6)H(5) and Atra also differentially affected the ability of eosin-5-maleimide (EMA) to alkylate Cys160 in the ANT protein and Atra, but not DIM-C-pPhtBu, inhibited the exchange of ATP/ADP in isolated mitochondria suggesting that these pharmacophores act on different sites on the ANT protein. Results of this study show that the receptor-independent proapoptotic activity of DIM-C-pPhtBu and DIM-C-pPhC(6)H(5) were related to novel mitochondriotoxic activities involving inner mitochondrial ANT proteins.
Collapse
Affiliation(s)
- Jun Hong
- Institute of Biosciences and Technology, Texas A&M University System Health Science Center, Houston, Texas 77843-4466, USA
| | | | | | | |
Collapse
|
334
|
Abstract
In the brain, glucocorticoids exert functions in neurogenesis, synaptic plasticity and behavioural responses, as well as in the control of hypothalamic-pituitary-adrenal axis activity. The generation of mice harbouring germline mutations that result either in loss or in gain of glucocorticoid receptor function provided a useful tool for understanding the role of glucocorticoids in the brain in vivo. The improvement of genomic technologies additionally allowed the establishment of mouse models with function-selective point mutations of the receptor as well as the generation of mice harbouring spatially and/or temporally restricted loss of glucocorticoid receptor, specifically within the brain. These models will provide the opportunity to better understand the mechanisms involved in glucocorticoid signalling within the nervous system.
Collapse
Affiliation(s)
- G Erdmann
- German Cancer Research Center, Division Molecular Biology of the Cell I, Im Neuenheimer Feld 280, Heidelberg, Germany
| | | | | |
Collapse
|
335
|
Flood L, Innala E, Löfberg R, Wikström AC. Patients with ulcerative colitis responding to steroid treatment up-regulate glucocorticoid receptor levels in colorectal mucosa. J Crohns Colitis 2008; 2:123-30. [PMID: 21172202 DOI: 10.1016/j.crohns.2007.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 10/17/2007] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Glucocorticosteroid treatment (GCS) is effective for attacks of ulcerative colitis (UC). However, 25-30% of patients fails to respond and may be considered steroid resistant. Glucocorticoid receptors (GR) mediate the effects of GCS. Colorectal mucosa levels of GR and NF-κB were analysed before, during and after treatment with GCS-compounds. METHODS Patients with moderate-severe attacks of ulcerative colitis were included. Patients undergoing colonoscopy with normal finding served as controls. GR and NF-κB levels in colorectal mucosa were analysed by Western Blotting and the DNA-binding activity of NF-κB by EMSA. RESULTS Twenty-eight patients and seven controls were included. Ten patients were judged clinically steroid resistant. Responders had significantly higher levels of GR in colorectal mucosa after one week of treatment than non-responders (P=0.039) and significantly higher levels of GR were found in responders in remission as compared to before treatment (P=0.013). NF-κB levels did not differ between the groups at first visit. Increasing levels were found only in responders as remission was obtained (P=0.031). EMSA detected 20% lower DNA-binding of NF-κB in responders in remission as compared to first visit (P=0.021). CONCLUSION GR levels increase in UC-patients responding to GCS-therapy but not in steroid resistant patients and may be the reason for the lack of steroid-efficacy. Increasing NF-κB levels were found in responders attaining remission, possibly reflecting a lower turnover. A decrease in DNA-binding of NF-κB was found in these patients, perhaps because of the increased GR levels counteracting NF-κB activity.
Collapse
Affiliation(s)
- L Flood
- Department of Gastroenterology and Hepatology, Novum, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden; IBD-Unit, HMQ Sophia Hospital, Solna, Stockholm, Sweden
| | | | | | | |
Collapse
|
336
|
Wang Y, Li T, Qiu X, Mo X, Zhang Y, Song Q, Ma D, Han W. CMTM3 can affect the transcription activity of androgen receptor and inhibit the expression level of PSA in LNCaP cells. Biochem Biophys Res Commun 2008; 371:54-8. [DOI: 10.1016/j.bbrc.2008.03.143] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 03/31/2008] [Indexed: 11/17/2022]
|
337
|
Wagner AE, Huck G, Stiehl DP, Jelkmann W, Hellwig-Bürgel T. Dexamethasone impairs hypoxia-inducible factor-1 function. Biochem Biophys Res Commun 2008; 372:336-40. [PMID: 18501194 DOI: 10.1016/j.bbrc.2008.05.061] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Accepted: 05/09/2008] [Indexed: 01/22/2023]
Abstract
Hypoxia-inducible factor-1 (HIF-1) is a heterodimeric transcription-factor composed of alpha- and beta-subunits. HIF-1 is not only necessary for the cellular adaptation to hypoxia, but it is also involved in inflammatory processes and wound healing. Glucocorticoids (GC) are therapeutically used to suppress inflammatory responses. Herein, we investigated whether GC modulate HIF-1 function using GC receptor (GR) possessing (HepG2) and GR deficient (Hep3B) human hepatoma cell cultures as model systems. Dexamethasone (DEX) treatment increased HIF-1alpha levels in the cytosol of HepG2 cells, while nuclear HIF-1alpha levels and HIF-1 DNA-binding was reduced. In addition, DEX dose-dependently lowered the hypoxia-induced luciferase activity in a reporter gene system. DEX suppressed the hypoxic stimulation of the expression of the HIF-1 target gene VEGF (vascular endothelial growth factor) in HepG2 cultures. DEX did not reduce hypoxically induced luciferase activity in HRB5 cells, a Hep3B derivative lacking GR. Transient expression of the GR in HRB5 cells restored the susceptibility to DEX. Our study discloses the inhibitory action of GC on HIF-1 dependent gene expression, which may be important with respect to the impaired wound healing in DEX-treated patients.
Collapse
Affiliation(s)
- A E Wagner
- Institute of Physiology, University of Luebeck, Ratzeburger Allee 160, D-23538 Luebeck, Germany
| | | | | | | | | |
Collapse
|
338
|
Haller J, Mikics E, Makara GB. The effects of non-genomic glucocorticoid mechanisms on bodily functions and the central neural system. A critical evaluation of findings. Front Neuroendocrinol 2008; 29:273-91. [PMID: 18054070 DOI: 10.1016/j.yfrne.2007.10.004] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 10/02/2007] [Accepted: 10/17/2007] [Indexed: 11/15/2022]
Abstract
Mounting evidence suggests that--beyond the well-known genomic effects--glucocorticoids affect cell function via non-genomic mechanisms. Such mechanisms operate in many major systems and organs including the cardiovascular, immune, endocrine and nervous systems, smooth and skeletal muscles, liver, and fat cells. Non-genomic effects are exerted by direct actions on membrane lipids (affecting membrane fluidity), membrane proteins (e.g. ion channels and neurotransmitter receptors), and cytoplasmic proteins (e.g. MAPKs, phospholipases, protein kinases, etc.). These actions are mediated by the glucocorticoids per se or by the proteins dissociated from the liganded glucocorticoid receptor complex. The MR and GR also activate non-genomic mechanisms in certain cases. Some effects of glucocorticoids are shared by a variety of steroids, whereas others are more selective. Moreover, "ultra-selective" effects-mediated by certain glucocorticoids only-were also shown. Disparate findings suggest that non-genomic mechanisms also show "demand-specificity", i.e. require the coincidence of two or more processes. Some of the non-genomic mechanisms activated by glucocorticoids are therapeutically relevant; moreover, the "non-genomic specificity" of certain glucocorticoids raises the possibility of therapeutic applications. Despite the large body of evidence, however, the non-genomic mechanisms of glucocorticoids are still poorly understood. Criteria for differentiating genomic and non-genomic mechanisms are often loosely applied; interactions between various mechanisms are unknown, and non-genomic mechanism-specific pharmacological (potentially therapeutic) agents are lacking. Nevertheless, the discovery of non-genomic mechanisms is a major breakthrough in stress research, and further insights into these mechanisms may open novel approaches for the therapy of various diseases.
Collapse
Affiliation(s)
- József Haller
- Department of Behavioral Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest 1083, Hungary
| | | | | |
Collapse
|
339
|
Yokoyama K, Hayashi M, Mogi C, Sasakawa Y, Watanabe G, Taya K, Devnath S, Inoue K. Dose-dependent effects of a glucocorticoid on prolactin production. Endocr J 2008; 55:405-14. [PMID: 18379123 DOI: 10.1507/endocrj.k07e-063] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Glucocorticoids are known to stimulate growth hormone (GH) production but to suppress prolactin (PRL) production. However, previous data were obtained with rather high doses of corticosterone. In this study we examined the effects of various doses (10 (-12) -10 (-7) M) of corticosterone on GH and PRL production in a rat pituitary somatomammotropic cell line, MtT/SM cells, and found that GH mRNA expression was facilitated by high doses (10 (-7) and 10 (-8) M). In contrast, a biphasic effect of corticosterone on PRL mRNA expression and secretion was observed, i.e., high doses (10 (-7) and 10 (-8) M) suppressed and low doses (10 (-12) -10 (-10) M) facilitated them. In an immunofluorescent staining study, the number of PRL immunopositive cells increased with low doses of corticosterone while it decreased with high doses of it, which corresponded to PRL mRNA expression and hormone secretion, respectively. These effects of corticosterone on PRL production were abolished by a glucocorticoid receptor (GR) antagonist, mifepristone. In addition, co-treatment with low doses of corticosterone (10 (-12) -10 (-10) M) and 17beta-estradiol (E(2), 10 nM) additively increased the number of PRL immunopositive cells. Moreover, a 24 h BrdU incorporation experiment suggested that the increase in the number of PRL immunopositive cells treated with low dose corticosterone was caused by novel synthesis of PRL while, on the other hand, that of those treated with E(2) resulted from PRL cell proliferation. Thus, we concluded that corticosterone biphasically regulates PRL production and the sensitivity of E(2) to different degrees.
Collapse
Affiliation(s)
- Kotaro Yokoyama
- Graduate School of Science and Engineering, Saitama University, Sakura-ku, Saitama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
340
|
Lin Z, Shen H, Huang J, Chen S, Chen L, Chen J, Liu G, Jiang H, Shen X. Butyl 4-(butyryloxy)benzoate functions as a new selective estrogen receptor beta agonist and induces GLUT4 expression in CHO-K1 cells. J Steroid Biochem Mol Biol 2008; 110:150-6. [PMID: 18455388 DOI: 10.1016/j.jsbmb.2008.03.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2007] [Revised: 02/18/2008] [Accepted: 03/20/2008] [Indexed: 11/29/2022]
Abstract
Estrogen receptors (ERs) belong to nuclear hormone receptor superfamily and can be activated by estrogens and regulate many target genes. Two ER isoforms, ERalpha and ERbeta have been discovered to date and ERbeta was indicated to involve in anti-inflammatory and anti-diabetogenic effects. Recently, some studies also demonstrated an association between ERbeta and GLUT4 expression. The development of selective ERbeta ligand has facilitated probing its novel biological functions and clinical benefits. In this work, a new ERbeta selective agonist, butyl 4-(butyryloxy)benzoate (DCW234), was discovered as investigated by surface plasmon resonance (SPR) technology, yeast two-hybrid and cell-based transcription-based assays. SPR results demonstrated that DCW234 has a higher binding affinity against ERbeta over ERalpha and induces a strong and selective stimulation on ERbeta/SRC1 interaction, which could be efficiently blocked by Tamoxifen. Meanwhile, the yeast two-hybrid technology-based assay indicated that DCW234 exhibits a higher agonistic activity ( approximately 13-fold) in stimulating ERbeta ligand-binding domain (LBD) interaction with SRC1 (EC(50)=2.5 microM) than ERalpha-LBD/SRC1 interaction (EC(50)=32.7 microM). The cell-based transcriptional assay further proved the potency and selectivity of DCW234. Moreover, DCW234 was found to be able to induce GLUT4 expression in CHO-K1 cell. The discovered DCW234 might be hopefully developed as a potential lead compound for further research.
Collapse
Affiliation(s)
- Zhaohu Lin
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | | | | | | | | | | | |
Collapse
|
341
|
Mukaide H, Adachi Y, Taketani S, Iwasaki M, Koike-Kiriyama N, Shigematsu A, Shi M, Yanai S, Yoshioka K, Kamiyama Y, Ikehara S. FKBP51 expressed by both normal epithelial cells and adenocarcinoma of colon suppresses proliferation of colorectal adenocarcinoma. Cancer Invest 2008; 26:385-390. [PMID: 18443959 DOI: 10.1080/07357900701799228] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
It has been reported, as a result of Western blot analyses, that FKBP51 is expressed in various tissues, but that it is not expressed in the pancreas, lung, colon, stomach, or spleen. In this paper, we show, using Western blot analyses, reverse transcriptase polymerase chain reaction, and immunohistochemical analyses of samples from colon cancer patients, that both normal epithelial cells and adenocarcinoma in the human colon express FKBP51, and that there are no significant differences in the expressions of FKBP51 between them. We also show that FKBP51 suppresses the proliferation of colorectal adenocarcinoma, possibly due to the suppression of functions of the glucocorticoid receptors.
Collapse
Affiliation(s)
- Hiromi Mukaide
- First Department of Pathology, Kansai Medical University, Moriguchi City, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
342
|
Zhang X, Clark AF, Yorio T. FK506-binding protein 51 regulates nuclear transport of the glucocorticoid receptor beta and glucocorticoid responsiveness. Invest Ophthalmol Vis Sci 2008; 49:1037-47. [PMID: 18326728 DOI: 10.1167/iovs.07-1279] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE A spliced variant of the human glucocorticoid receptor GRbeta has been implicated in glucocorticoid responsiveness in glaucoma. Over-expression of the FK506-binding immunophilin FKBP51 also causes a generalized state of glucocorticoid resistance. In the present study, the roles of FKBP51 in the nuclear transport of GRbeta and glucocorticoid responsiveness were investigated. METHODS Human trabecular meshwork cells (GTM3 and TM5) and HeLa cells were treated with dexamethasone (DEX) and FK506 and transfected with GRbeta and FKBP51 expression vectors. Coimmunoprecipitation and Western blot analyses were performed to study interactions of FKBP51 and FKBP52 with GRalpha, GRbeta, Hsp90, or dynein. The cells were transfected with a GRE-luciferase reporter to evaluate the effects of DEX and FK506 and the overexpression of GRbeta and FKBP51 on glucocorticoid-mediated gene expression. RESULTS FKBP51 was involved in constitutive nuclear transport of both GRalpha and -beta in the absence of ligands. FKBP52 appeared to be solely responsible for the nuclear transport of ligand-activated GRalpha. DEX stimulated the translocation of GRalpha but not GRbeta. Overexpression of either GRbeta or FKBP51 stimulated GRbeta translocation and reduced DEX-induced luciferase in HeLa cells. FK506 did not alter DEX-induced translocation of GRalpha. However, FK506 increased the association of FKBP51 with GRbeta and stimulated DEX-induced translocation of GRbeta in normal TM cells, but not in glaucoma TM cells. Increased nuclear GRbeta significantly inhibited glucocorticoid responsiveness in TM cells. CONCLUSIONS Nuclear transport of GRbeta represents a novel mechanism through which FKBP51 alters GC sensitivity. GRbeta and FKBP51 may be responsible for increased responsiveness in steroid-induced ocular hypertensive individuals as well as in patients with glaucoma.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107, USA
| | | | | |
Collapse
|
343
|
A SIM-ultaneous role for SUMO and ubiquitin. Trends Biochem Sci 2008; 33:201-8. [PMID: 18403209 DOI: 10.1016/j.tibs.2008.02.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 02/08/2008] [Accepted: 02/12/2008] [Indexed: 11/22/2022]
Abstract
Ubiquitin and ubiquitin-like proteins (Ubls) share a beta-GRASP fold and have key roles in cellular growth and suppression of genome instability. Despite their common fold, SUMO and ubiquitin are classically portrayed as distinct, and they can have antagonistic roles. Recently, a new family of proteins, the small ubiquitin-related modifier (SUMO)-targeted ubiquitin ligases (STUbLs), which directly connect sumoylation and ubiquitylation, has been discovered. Uniquely, STUbLs use SUMO-interaction motifs (SIMs) to recognize their sumoylated targets. STUbLs are global regulators of protein sumoylation levels, and cells lacking STUbLs display genomic instability and hypersensitivity to genotoxic stress. The human STUbL, RNF4, is implicated in several diseases including cancer, highlighting the importance of characterizing the cellular functions of STUbLs.
Collapse
|
344
|
Rocha Viegas L, Hoijman E, Beato M, Pecci A. Mechanisms involved in tissue-specific apopotosis regulated by glucocorticoids. J Steroid Biochem Mol Biol 2008; 109:273-8. [PMID: 18424036 DOI: 10.1016/j.jsbmb.2008.03.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Physiological cell turnover is under the control of a sharp and dynamic balance of different homeostatic mechanisms such as the equilibrium between cell proliferation and cell death. These mechanisms play an important role in maintaining normal tissue function and architecture. It is well known that apoptosis is the prevalent mode of physiological cell loss in most tissues. Steroid hormones like glucocorticoids have been identified as key signals controlling cell turnover by modulating programmed cell death in a tissue- and cell-specific manner. In this sense, several reports have demonstrated that glucocorticoids are able to induce apoptosis in cells of the hematopoietic system such as monocytes, macrophages, and T lymphocytes. In contrast, they protect against apoptotic signals evoked by cytokines, cAMP, tumor suppressors, in glandular cells such as the mammary gland epithelia, endometrium, hepatocytes, ovarian follicular cells, and fibroblasts. Although several studies have provided significant information on hormone-dependent apoptosis in an specific tissue, a clearly defined pathway that mediates cell death in response to glucocorticoids in different cell types is still misunderstood. The scope of this review is held to those mechanisms by which glucocorticoids control apoptosis, emphasizing tissue-specific expression of genes that are involved in the apoptotic pathway.
Collapse
Affiliation(s)
- Luciana Rocha Viegas
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Cdad. Universitaria, Pab. II, C1428EGA Buenos Aires, Argentina
| | | | | | | |
Collapse
|
345
|
Abstract
Steroid hormones working through their receptors regulate a wide variety of physiologic processes necessary for normal homeostasis. Recent years have witnessed great advances in our understanding of how these hormones interact with their receptors, and have brought us closer to the era of directed drug design. We previously described a novel intramolecular interaction between helix 3 and helix 5 which is responsible for a Mendelian form of human hypertension. Further studies revealed that this interaction is highly conserved throughout the steroid hormone receptor family and functions as a key regulator of steroid hormone receptor sensitivity and specificity. Here, we review the contribution of helix 3-helix 5 interaction to steroid hormone receptor activity, with an eye towards how this knowledge may aid in the creation of novel therapeutic agonists and antagonists.
Collapse
Affiliation(s)
- Junhui Zhang
- Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520-8029, USA
| | | |
Collapse
|
346
|
Thomas RS, Sarwar N, Phoenix F, Coombes RC, Ali S. Phosphorylation at serines 104 and 106 by Erk1/2 MAPK is important for estrogen receptor-alpha activity. J Mol Endocrinol 2008; 40:173-84. [PMID: 18372406 PMCID: PMC2277492 DOI: 10.1677/jme-07-0165] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Phosphorylation of estrogen receptor-alpha (ERalpha) at specific residues in transcription activation function 1 (AF-1) can stimulate ERalpha activity in a ligand-independent manner. This has led to the proposal that AF-1 phosphorylation and the consequent increase in ERalpha activity could contribute to resistance to endocrine therapies in breast cancer patients. Previous studies have shown that serine 118 (S118) in AF-1 is phosphorylated by extracellular signal-regulated kinases 1 and 2 (Erk1/2) mitogen-activated protein kinase (MAPK) in a ligand-independent manner. Here, we show that serines 104 (S104) and 106 (S106) are also phosphorylated by MAPK in vitro and upon stimulation of MAPK activity in vivo. Phosphorylation of S104 and S106 can be inhibited by the MAP-erk kinase (MEK)1/2 inhibitor U0126 and by expression of kinase-dead Raf1. Further, we show that, although S118 is important for the stimulation of ERalpha activity by the selective ER modulator 4-hydroxytamoxifen (OHT), S104 and S106 are also required for the agonist activity of OHT. Acidic amino acid substitution of S104 or S106 stimulates ERalpha activity to a greater extent than the equivalent substitution at S118, suggesting that phosphorylation at S104 and S106 is important for ERalpha activity. Collectively, these data indicate that the MAPK stimulation of ERalpha activity involves the phosphorylation not only of S118 but also of S104 and S106, and that MAPK-mediated hyperphosphorylation of ERalpha at these sites may contribute to resistance to tamoxifen in breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Simak Ali
- (Correspondence should be addressed to S Ali; )
| |
Collapse
|
347
|
Progesterone induction of the 11beta-hydroxysteroid dehydrogenase type 2 promoter in breast cancer cells involves coordinated recruitment of STAT5A and progesterone receptor to a distal enhancer and polymerase tracking. Mol Cell Biol 2008; 28:3830-49. [PMID: 18378698 DOI: 10.1128/mcb.01217-07] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Steroid hormone receptors regulate gene expression, interacting with target DNA sequences but also activating cytoplasmic signaling pathways. Using the human 11beta-hydroxysteroid dehydrogenase type 2 (11beta-HSD2) gene as a model, we have investigated the contributions of both effects on a human progesterone-responsive promoter in breast cancer cells. Chromatin immunoprecipitation has identified two different mechanisms of hormone-induced progesterone receptor (PR) recruitment to the 11beta-HSD2 promoter: (i) direct PR binding to DNA at the proximal promoter, abrogated when PR contains a mutated DNA binding domain (DBD), and (ii) STAT5A (signal transducer and activator of transcription 5A)-mediated recruitment of PR to an upstream distal region, impaired by AG490, a JAK/STAT pathway inhibitor. The JAK/STAT inhibitor, as well as expression of dominant-negative STAT5A, impairs hormone induction of 11beta-HSD2. On the other hand, the DBD-mutated PR fully supports 11beta-HSD2 expression. These results, along with data from a deletion analysis, indicate that the distal region is crucial for hormone regulation of 11beta-HSD2. We show active RNA polymerase II tracking from the distal region upon PR and STAT5A binding, concomitant with synthesis of noncoding, hormone-dependent RNAs, suggesting that this region works as a hormone-dependent transcriptional enhancer. In conclusion, coordination of PR transcriptional effects and cytoplasmic signaling activation, in particular the JAK/STAT pathway, are critical in regulating progestin-induced endogenous 11beta-HSD2 gene expression in breast cancer cells. This is not unique to this promoter, as AG490 also alters the expression of other progesterone-regulated genes.
Collapse
|
348
|
Ho KJ, Liao JK. Non-nuclear actions of estrogen: new targets for prevention and treatment of cardiovascular disease. Mol Interv 2008; 2:219-28. [PMID: 14993393 PMCID: PMC2633129 DOI: 10.1124/mi.2.4.219] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Gender-based differences in the incidence of hypertensive and coronary artery disease, the development of atherosclerosis, and myocardial remodeling after infarction are attributable to the indirect effect of estrogen on risk factor profiles, such as cholesterol levels, glucose metabolism, and insulin levels. More recent evidence, however, suggests that activated estrogen receptor (ER) mediates signaling cascades that culminate in direct protective effects such as vasodilation, inhibition of response to vessel injury, limiting myocardial injury after infarction, and attenuating cardiac hypertrophy. Although the ER is usually thought of as a ligand-dependent transcription factor, it can also rapidly mobilize signals at the plasma membrane and in the cytoplasm. Thus, a greater understanding of ER function and regulation may lead to the development of highly specific therapeutics that mediate the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Karen J Ho
- The Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Cambridge, MA 02139, USA
| | | |
Collapse
|
349
|
Fischer B, Fukuzawa K, Wenzel W. Receptor-specific scoring functions derived from quantum chemical models improve affinity estimates for in-silico drug discovery. Proteins 2008; 70:1264-73. [PMID: 17876816 DOI: 10.1002/prot.21607] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The adaptation of forcefield-based scoring function to specific receptors remains an important challenge for in-silico drug discovery. Here we compare binding energies of forcefield-based scoring functions with models that are reparameterized on the basis of large-scale quantum calculations of the receptor. We compute binding energies of eleven ligands to the human estrogen receptor subtype alpha (ERalpha) and four ligands to the human retinoic acid receptor of isotype gamma (RARgamma). Using the FlexScreen all-atom receptor-ligand docking approach, we compare docking simulations parameterized by quantum-mechanical calculation of a large protein fragment with purely forcefield-based models. The use of receptor flexibility in the FlexScreen permits the treatment of all ligands in the same receptor model. We find a high correlation between the classical binding energy obtained in the docking simulation and quantum mechanical binding energies and a good correlation with experimental affinities R=0.81 for ERalpha and R=0.95 for RARgamma using the quantum derived scoring functions. A significant part of this improvement is retained, when only the receptor is treated with quantum-based parameters, while the ligands are parameterized with a purely classical model.
Collapse
Affiliation(s)
- Bernhard Fischer
- Forschungszentrum Karlsruhe, Institut für Nanotechnologie, Postfach 3640, D-76021 Karlsruhe, Germany
| | | | | |
Collapse
|
350
|
Suino-Powell K, Xu Y, Zhang C, Tao YG, Tolbert WD, Simons SS, Xu HE. Doubling the size of the glucocorticoid receptor ligand binding pocket by deacylcortivazol. Mol Cell Biol 2008; 28:1915-23. [PMID: 18160712 PMCID: PMC2268401 DOI: 10.1128/mcb.01541-07] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 10/10/2007] [Accepted: 12/17/2007] [Indexed: 01/14/2023] Open
Abstract
A common feature of nuclear receptor ligand binding domains (LBD) is a helical sandwich fold that nests a ligand binding pocket within the bottom half of the domain. Here we report that the ligand pocket of glucocorticoid receptor (GR) can be continuously extended into the top half of the LBD by binding to deacylcortivazol (DAC), an extremely potent glucocorticoid. It has been puzzling for decades why DAC, which contains a phenylpyrazole replacement at the conserved 3-ketone of steroid hormones that are normally required for activation of their cognate receptors, is a potent GR activator. The crystal structure of the GR LBD bound to DAC and the fourth LXXLL motif of steroid receptor coactivator 1 reveals that the GR ligand binding pocket is expanded to a size of 1,070 A(3), effectively doubling the size of the GR dexamethasone-binding pocket of 540 A(3) and yet leaving the structure of the coactivator binding site intact. DAC occupies only approximately 50% of the space of the pocket but makes intricate interactions with the receptor around the phenylpyrazole group that accounts for the high-affinity binding of DAC. The dramatic expansion of the DAC-binding pocket thus highlights the conformational adaptability of GR to ligand binding. The new structure also allows docking of various nonsteroidal ligands that cannot be fitted into the previous structures, thus providing a new rational template for drug discovery of steroidal and nonsteroidal glucocorticoids that can be specifically designed to reach the unoccupied space of the expanded pocket.
Collapse
Affiliation(s)
- Kelly Suino-Powell
- Laboratory of Structural Sciences, Van Andel Research Institute, 333 Bostwick Avenue, Grand Rapids, MI 49503, USA
| | | | | | | | | | | | | |
Collapse
|