151
|
Ikeda K, Horie-Inoue K, Inoue S. Analysis of TFRNs associated with steroid hormone-related cancers. Methods Mol Biol 2014; 1164:197-209. [PMID: 24927845 DOI: 10.1007/978-1-4939-0805-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Estrogen and androgen are important endocrine hormones that control physiological functions in reproductive organs and play roles in the generation and/or progression of steroid hormone-related cancers. Their cognate receptors function as hormone-dependent transcription factors, which directly regulate the expression of their target genes. Genome-wide analysis of hormone receptor-related networks will provide new insights into the understanding of the molecular mechanism orchestrated by estrogen and androgen receptors, and will enable the development of new methods for the diagnosis and treatment of steroid hormone-related cancers.
Collapse
Affiliation(s)
- Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, 1397-1 Yamane, Hidaka, Saitama, 350-1241, Japan
| | | | | |
Collapse
|
152
|
Abstract
Understanding how eukaryotic gene regulation works implies unraveling the mechanisms used by transcription factors to access DNA information packaged in chromatin. The current view is that different cell types express different parts of the genome because they are equipped with different sets of transcription factors. A few transcription factors are called pioneer factors because they are able to bind to their sites in nucleosomes and to open up chromatin thus enabling access for other transcription factors, which are unable to recognize DNA packaged in nucleosomes. But it is also possible that the way DNA is organized in chromatin differs between cell types and contributes to cell identity by restricting or enhancing access to specific gene cohorts. To unravel these mechanisms we studied the interaction of progesterone receptor with the genome of breast cancer cells and found that it binds preferentially to sites organized in nucleosomes, which contribute to functional interactions leading to gene regulation.
Collapse
|
153
|
Kuzmich D, Bentzien J, Betageri R, DiSalvo D, Fadra-Khan T, Harcken C, Kukulka A, Nabozny G, Nelson R, Pack E, Souza D, Thomson D. Function-regulating pharmacophores in a sulfonamide class of glucocorticoid receptor agonists. Bioorg Med Chem Lett 2013; 23:6640-4. [DOI: 10.1016/j.bmcl.2013.10.052] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 11/16/2022]
|
154
|
Substituted phenyl as a steroid A-ring mimetic: Providing agonist activity to a class of arylsulfonamide nonsteroidal glucocorticoid ligands. Bioorg Med Chem Lett 2013; 23:6645-9. [DOI: 10.1016/j.bmcl.2013.10.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/22/2013] [Accepted: 10/23/2013] [Indexed: 01/24/2023]
|
155
|
McDermott GP, Do D, Litterst CM, Maar D, Hindson CM, Steenblock ER, Legler TC, Jouvenot Y, Marrs SH, Bemis A, Shah P, Wong J, Wang S, Sally D, Javier L, Dinio T, Han C, Brackbill TP, Hodges SP, Ling Y, Klitgord N, Carman GJ, Berman JR, Koehler RT, Hiddessen AL, Walse P, Bousse L, Tzonev S, Hefner E, Hindson BJ, Cauly TH, Hamby K, Patel VP, Regan JF, Wyatt PW, Karlin-Neumann GA, Stumbo DP, Lowe AJ. Multiplexed Target Detection Using DNA-Binding Dye Chemistry in Droplet Digital PCR. Anal Chem 2013; 85:11619-27. [PMID: 24180464 DOI: 10.1021/ac403061n] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Geoffrey P. McDermott
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Duc Do
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Claudia M. Litterst
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Dianna Maar
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | | | - Erin R. Steenblock
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Tina C. Legler
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Yann Jouvenot
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Samuel H. Marrs
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Adam Bemis
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Pallavi Shah
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Josephine Wong
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Shenglong Wang
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - David Sally
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Leanne Javier
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Theresa Dinio
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Chunxiao Han
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Timothy P. Brackbill
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Shawn P. Hodges
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Yunfeng Ling
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Niels Klitgord
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - George J. Carman
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Jennifer R. Berman
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Ryan T. Koehler
- VerdAscend Sciences, West Linn, Oregon, 97068, United States
| | - Amy L. Hiddessen
- Purigen Biosystems, Inc., Alviso, California 95002, United States
| | - Pramod Walse
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Luc Bousse
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Svilen Tzonev
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Eli Hefner
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | | | - Thomas H. Cauly
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Keith Hamby
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Viresh P. Patel
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - John F. Regan
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - Paul W. Wyatt
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | - George A. Karlin-Neumann
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| | | | - Adam J. Lowe
- The
Digital Biology Center, Bio-Rad Laboratories, Inc., 5731 West Las Positas
Boulevard, Pleasanton, California 94566, United States
| |
Collapse
|
156
|
Balogh P, Szabó A, Katz S, Likó I, Patócs A, L.Kiss A. Estrogen receptor alpha is expressed in mesenteric mesothelial cells and is internalized in caveolae upon Freund's adjuvant treatment. PLoS One 2013; 8:e79508. [PMID: 24244516 PMCID: PMC3828353 DOI: 10.1371/journal.pone.0079508] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 09/27/2013] [Indexed: 12/03/2022] Open
Abstract
Transformation of epithelial cells into connective tissue cells (epithelial-mesenchymal transition, EMT) is a complex mechanism involved in tumor metastasis, and in normal embryogenesis, while type II EMT is mainly associated with inflammatory events and tissue regenaration. In this study we examined type II EMT at the ultrastructural and molecular level during the inflammatory process induced by Freund's adjuvant treatment in rat mesenteric mesothelial cells. We found that upon the inflammatory stimulus mesothelial cells lost contact with the basal lamina and with each other, and were transformed into spindle-shaped cells. These morphological changes were accompanied by release of interleukins IL-1alpha, -1beta and IL-6 and by secretion of transforming growth factor beta (TGF-β) into the peritoneal cavity. Mesothelial cells also expressed estrogen receptor alpha (ER-α) as shown by immunolabeling at the light and electron microscopical levels, as well as by quantitative RT-PCR. The mRNA level of ER-α showed an inverse correlation with the secretion of TGF-β. At the cellular and subcellular levels ER-α was colocalized with the coat protein caveolin-1 and was found in the plasma membrane of mesothelial cells, in caveolae close to multivesicular bodies (MVBs) or in the membrane of these organelles, suggesting that ER-α is internalized via caveola-mediated endocytosis during inflammation. We found asymmetric, thickened, electron dense areas on the limiting membrane of MVBs (MVB plaques) indicating that these sites may serve as platforms for collecting and organizing regulatory proteins. Our morphological observations and biochemical data can contribute to form a potential model whereby ER-α and its caveola-mediated endocytosis might play role in TGF-β induced type II EMT in vivo.
Collapse
Affiliation(s)
- Petra Balogh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
- * E-mail:
| | - Arnold Szabó
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Sándor Katz
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - István Likó
- Pharmacology and Drug Safety Research, R. Gedeon Plc, Hungary
| | - Attila Patócs
- HSA-SE Lendület Hereditary Endocrine Tumors Research Group, Budapest, Hungary
| | - Anna L.Kiss
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
157
|
Planey SL, Kumar R, Arnott JA. Estrogen receptors (ERαversus ERβ): friends or foes in human biology? J Recept Signal Transduct Res 2013; 34:1-5. [DOI: 10.3109/10799893.2013.853188] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
158
|
Janeczko A, Oklešťková J, Siwek A, Dziurka M, Pociecha E, Kocurek M, Novák O. Endogenous progesterone and its cellular binding sites in wheat exposed to drought stress. J Steroid Biochem Mol Biol 2013; 138:384-94. [PMID: 23973943 DOI: 10.1016/j.jsbmb.2013.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/24/2013] [Accepted: 07/29/2013] [Indexed: 12/24/2022]
Abstract
Progesterone is a basic hormone that regulates the metabolism in mammals. The presence of this compound has also been found in certain plants. It is believed that progesterone can regulate growth processes and resistance to stress, however, its precise role in plants remains unknown. The research conducted in this study was aimed at analyzing the content of endogenous progesterone and its cellular binding sites in the leaves of spring wheat exposed to drought. Changes were studied in two cultivars of wheat - a cultivar sensitive to drought (Katoda) and tolerant cultivar (Monsun). Plants had undergone periodic droughts during the seedling stage or in the phase of heading. The occurrence of free progesterone as well as its conjugated forms was observed in wheat studied. The amount of progesterone ranged from 0.2 to 5.8pmolgFW(-1) and was dependent on the cultivar, age of the plants, stage of development and fluctuated as a result of the exposure to drought. Cv. Katoda responded to a water deficit by lowering the amount of progesterone and cv. Monsun by increasing its level. Progesterone in plants grown in limited water conditions occurred primarily in a free form. While in the optimal watering conditions, some of its pool was found in the form of conjugates. In the spring wheat the occurrence of binding sites for progesterone was detected in cell membranes, cytoplasm and nuclei in the range of 10-36fmol/mg of protein. The wheat cultivars tested, Monsun and Katoda, differ in their concentration of cellular binding sites for progesterone. This number varied in the individual fractions during different stages of plant development and due to the effect of drought stress. The number of binding sites for progesterone located in the membrane fraction of seedlings and flag leaves increased significantly under drought in the cv. Katoda (35-46%), but did not change in the cv. Monsun. Whereas the number of cytoplasmic progesterone binding sites increased during the drought in the cv. Monsun (about 50%), they did not change in the cv. Katoda. Changes in the amount of progesterone and its binding sites in the cell under the influence of drought were then different depending on whether the cultivar was tolerant or sensitive to drought. The possibility of utilizing these changes as markers of drought resistance is discussed. The results obtained suggest that progesterone is a part of wheat response to stress factors (drought).
Collapse
Affiliation(s)
- Anna Janeczko
- Institute of Plant Physiology, Polish Academy of Sciences, Niezapominajek 21, 30-239 Krakow, Poland.
| | | | | | | | | | | | | |
Collapse
|
159
|
Vasiljević A, Veličković N, Bursać B, Djordjevic A, Milutinović DV, Nestorović N, Matić G. Enhanced prereceptor glucocorticoid metabolism and lipogenesis impair insulin signaling in the liver of fructose-fed rats. J Nutr Biochem 2013; 24:1790-7. [DOI: 10.1016/j.jnutbio.2013.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/18/2013] [Accepted: 04/01/2013] [Indexed: 12/19/2022]
|
160
|
Piccinato CA, Rosa GJM, N’Jai AU, Jefcoate CR, Wiltbank MC. Estradiol and progesterone exhibit similar patterns of hepatic gene expression regulation in the bovine model. PLoS One 2013; 8:e73552. [PMID: 24069207 PMCID: PMC3775788 DOI: 10.1371/journal.pone.0073552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Female sex steroid hormones, estradiol-17β (E2-17β) and progesterone (P4) regulate reproductive function and gene expression in a broad range of tissues. Given the central role of the liver in regulating homeostasis including steroid hormone metabolism, we sought to understand how E2-17β and P4 interact to affect global gene expression in liver. Ovariectomized cows (n = 8) were randomly assigned to 4 treatment groups applied in a replicated Latin Square design: 1) No hormone supplementation, 2) E2-17β treatment (ear implant), 3) P4 treatment (intravaginal inserts), and 4) E2-17β combined with P4. After 14 d of treatment, liver biopsies were collected, allowing 28 d intervals between periods. Changes in gene expression in the liver biopsies were monitored using bovine-specific arrays. Treatment with E2-17β altered expression of 479 genes, P4 472 genes, and combined treatment significantly altered expression of 468 genes. In total, 578 genes exhibited altered expression including a remarkable number (346 genes) that responded similarly to E2-17β, P4, or combined treatment. Additional evidence for similar gene expression actions of E2-17ß and/or P4 were: principal component analysis placed almost every treatment array at a substantial distance from controls; Venn diagrams indicated overall treatment effects for most regulated genes; clustering analysis indicated the two major clusters had all treatments up-regulating (172 genes) or down-regulating (173 genes) expression. Thus, unexpectedly, common biological pathways were regulated by E2-17β and/or P4 in liver. This indicates that the mechanism of action of these steroid hormones in the liver might be either indirect or might occur through non-genomic pathways. This unusual pattern of gene expression in response to steroid hormones is consistent with the idea that there are classical and non-classical tissue-specific responses to steroid hormone actions. Future studies are needed to elucidate putative mechanism(s) responsible for overlapping actions of E2-17β and P4 on the liver transcriptome.
Collapse
Affiliation(s)
- Carla A. Piccinato
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Guilherme J. M. Rosa
- Department of Animal Sciences, and Department of Biostatistics & Medical Informatics, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Alhaji U. N’Jai
- Department of Pathobiological Sciences and Molecular & Environmental Toxicology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Colin R. Jefcoate
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Pharmacology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Milo C. Wiltbank
- Endocrinology-Reproductive Physiology Program, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Dairy Science, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
161
|
Takigawa T, Miyazaki H, Kinoshita M, Kawarabayashi N, Nishiyama K, Hatsuse K, Ono S, Saitoh D, Seki S, Yamamoto J. Glucocorticoid receptor-dependent immunomodulatory effect of ursodeoxycholic acid on liver lymphocytes in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G427-38. [PMID: 23868404 DOI: 10.1152/ajpgi.00205.2012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Although ursodeoxycholic acid (UDCA) has long been used for patients with chronic cholestatic liver diseases, particularly primary biliary cirrhosis, it may modulate the host immune response. This study investigated the effect of UDCA feeding on experimental hepatitis, endotoxin shock, and bacterial infection in mice. C57BL/6 mice were fed a diet supplemented with or without 0.3% (wt/vol) UDCA for 4 wk. UDCA improved hepatocyte injury and survival in concanavalin-A (Con-A)-induced hepatitis by suppressing IFN-γ production by liver mononuclear cells (MNC), especially NK and NKT cells. UDCA also increased survival after lipopolysaccharide (LPS)-challenge; however, it increased mortality of mice following Escherichia coli infection due to the worsening of infection. UDCA-fed mice showed suppressed serum IL-18 levels and production of IL-18 from liver Kupffer cells, which together with IL-12 potently induce IFN-γ production. However, unlike normal mice, exogenous IL-18 pretreatment did not increase the serum IFN-γ levels after E. coli, LPS, or Con-A challenge in the UDCA-fed mice. Interestingly, however, glucocorticoid receptor (GR) expression was significantly upregulated in the liver MNC of the UDCA-fed mice but not in their whole liver tissue homogenates. Silencing GR in the liver MNC abrogated the suppressive effect of UDCA on LPS- or Con-A-induced IFN-γ production. Furthermore, RU486, a GR antagonist, restored the serum IFN-γ level in UDCA-fed mice after E. coli, LPS, or Con-A challenge. Taken together, these results suggest that IFN-γ-reducing immunomodulatory property of UDCA is mediated by elevated GR in the liver lymphocytes in an IL-12/18-independent manner.
Collapse
Affiliation(s)
- Toshimichi Takigawa
- Dept. of Immunology and Microbiology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513 Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Zhang X, Zhou Q, Chen L, Berger S, Wu H, Xiao Z, Pearce D, Zhou X, Zhang W. Mineralocorticoid receptor antagonizes Dot1a-Af9 complex to increase αENaC transcription. Am J Physiol Renal Physiol 2013; 305:F1436-44. [PMID: 24026182 DOI: 10.1152/ajprenal.00202.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aldosterone is a major regulator of Na(+) absorption and acts by activating the mineralocorticoid receptor (MR) to stimulate the epithelial Na(+) channel (ENaC). MR(-/-) mice exhibited pseudohypoaldosteronism type 1 (hyponatremia, hyperkalemia, salt wasting, and high levels of aldosterone) and died around postnatal day 10. However, if and how MR regulates ENaC transcription remain incompletely understood. Our earlier work demonstrated that aldosterone activates αENaC transcription by reducing expression of Dot1a and Af9 and by impairing Dot1a-Af9 interaction. Most recently, we reported identification of a major Af9 binding site in the αENaC promoter and upregulation of αENaC mRNA expression in mouse kidneys lacking Dot1a. Despite these findings, the putative antagonism between the MR/aldosterone and Dot1a-Af9 complexes has never been addressed. The molecular defects leading to PHA-1 in MR(-/-) mice remain elusive. Here, we report that MR competes with Dot1a to bind Af9. MR/aldosterone and Dot1a-Af9 complexes mutually counterbalance ENaC mRNA expression in inner medullary collecting duct 3 (IMCD3) cells. Real-time RT-quantitative PCR revealed that 5-day-old MR(-/-) vs. MR(+/+) mice had significantly lower αENaC mRNA levels. This change was associated with an increased Af9 binding and H3 K79 hypermethylation in the αENaC promoter. Therefore, this study identified MR as a novel binding partner and regulator of Af9 and a novel mechanism coupling MR-mediated activation with relief of Dot1a-Af9-mediated repression via MR-Af9 interaction. Impaired ENaC expression due to failure to inhibit Dot1a-Af9 may play an important role in the early stages of PHA-1 (before postnatal day 8) in MR(-/-) mice.
Collapse
Affiliation(s)
- Xi Zhang
- Dept. of Internal Medicine, Univ. of Texas Medical School at Houston, 6431 Fannin, MSB 5.135, Houston, TX 77030.
| | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Mannironi C, Camon J, De Vito F, Biundo A, De Stefano ME, Persiconi I, Bozzoni I, Fragapane P, Mele A, Presutti C. Acute stress alters amygdala microRNA miR-135a and miR-124 expression: inferences for corticosteroid dependent stress response. PLoS One 2013; 8:e73385. [PMID: 24023867 PMCID: PMC3762773 DOI: 10.1371/journal.pone.0073385] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 07/22/2013] [Indexed: 11/18/2022] Open
Abstract
The amygdala is a brain structure considered a key node for the regulation of neuroendocrine stress response. Stress-induced response in amygdala is accomplished through neurotransmitter activation and an alteration of gene expression. MicroRNAs (miRNAs) are important regulators of gene expression in the nervous system and are very well suited effectors of stress response for their ability to reversibly silence specific mRNAs. In order to study how acute stress affects miRNAs expression in amygdala we analyzed the miRNA profile after two hours of mouse restraint, by microarray analysis and reverse transcription real time PCR. We found that miR-135a and miR-124 were negatively regulated. Among in silico predicted targets we identified the mineralocorticoid receptor (MR) as a target of both miR-135a and miR-124. Luciferase experiments and endogenous protein expression analysis upon miRNA upregulation and inhibition allowed us to demonstrate that mir-135a and mir-124 are able to negatively affect the expression of the MR. The increased levels of the amygdala MR protein after two hours of restraint, that we analyzed by western blot, negatively correlate with miR-135a and miR-124 expression. These findings point to a role of miR-135a and miR-124 in acute stress as regulators of the MR, an important effector of early stress response.
Collapse
Affiliation(s)
- Cecilia Mannironi
- Institute of Cellular Biology and Neurobiology, Consiglio Nazionale delle Ricerche, A. Buzzati-Traverso Campus, Monterotondo, Rome, Italy
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
- * E-mail:
| | - Jeremy Camon
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
- Center for Research in Neurobiology, Sapienza University of Rome, Rome, Italy
| | - Francesca De Vito
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Antonio Biundo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Maria Egle De Stefano
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
- Center for Research in Neurobiology, Sapienza University of Rome, Rome, Italy
- Institute Pasteur Fondazione Cenci-Bolognetti, Rome, Italy
| | - Irene Persiconi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
- Institute Pasteur Fondazione Cenci-Bolognetti, Rome, Italy
| | - Irene Bozzoni
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| | - Paola Fragapane
- Institute of Molecular Biology and Pathology, Consiglio Nazionale delle Ricerche, Sapienza University of Rome, Rome, Italy
| | - Andrea Mele
- Institute of Cellular Biology and Neurobiology, Consiglio Nazionale delle Ricerche, A. Buzzati-Traverso Campus, Monterotondo, Rome, Italy
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
- Center for Research in Neurobiology, Sapienza University of Rome, Rome, Italy
| | - Carlo Presutti
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
164
|
Filippa V, Godoy D, Perez E, Mohamed F. Effects of castration on androgen receptors and gonadotropins in the pituitary of adult male viscachas. Reprod Fertil Dev 2013; 26:991-1000. [PMID: 23905557 DOI: 10.1071/rd13126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 06/26/2013] [Indexed: 11/23/2022] Open
Abstract
The aims of the present study were to determine whether castration results in quantitative immunohistochemical changes in androgen receptors (AR), LH-immunoreactive (IR) cells and FSH-IR cells, and to analyse the colocalisation of AR and gonadotropins in the pituitary pars distalis (PD) of viscachas. Pituitaries were processed for light and electron microscopy. AR-IR, LH-IR and FSH-IR cells were detected by immunohistochemistry. In morphometric studies, the percentage of AR-IR, LH-IR, FSH-IR, LH-IR/AR-IR and FSH-IR/AR-IR cells was determined. In intact viscachas, AR were distributed throughout the PD; they were numerous at the caudal end, with intense immunostaining. LH-IR cells and FSH-IR cells were found mainly in the ventral region and at the rostral end of the PD. Approximately 45%-66% of LH-IR cells and 49%-57% of FSH-IR cells expressed AR in the different zones of the PD. In castrated viscachas, there was a significant decrease in the percentage of AR-IR, LH-IR, FSH-IR, and FSH-IR/AR-IR cells. Some pituitary cells from castrated viscachas also exhibited ultrastructural changes. These results provide morphological evidence that gonadal androgens are directly related to the immunolabelling of AR, LH and FSH. Moreover, the colocalisation of AR and FSH is most affected by castration, suggesting the existence of a subpopulation of gonadotrophs with different regulatory mechanisms for hormonal synthesis, storage and secretion.
Collapse
Affiliation(s)
- Verónica Filippa
- Laboratorio de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Av. Ejército de los Andes 950- Bloque I, 1° Piso (5700) San Luis, Argentina
| | - Daiana Godoy
- Laboratorio de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Av. Ejército de los Andes 950- Bloque I, 1° Piso (5700) San Luis, Argentina
| | - Edith Perez
- Laboratorio de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Av. Ejército de los Andes 950- Bloque I, 1° Piso (5700) San Luis, Argentina
| | - Fabian Mohamed
- Laboratorio de Histología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Av. Ejército de los Andes 950- Bloque I, 1° Piso (5700) San Luis, Argentina
| |
Collapse
|
165
|
Kullmann MK, Grubbauer C, Goetsch K, Jäkel H, Podmirseg SR, Trockenbacher A, Ploner C, Cato ACB, Weiss C, Kofler R, Hengst L. The p27-Skp2 axis mediates glucocorticoid-induced cell cycle arrest in T-lymphoma cells. Cell Cycle 2013; 12:2625-35. [PMID: 23907123 PMCID: PMC3865052 DOI: 10.4161/cc.25622] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Glucocorticoid therapy is an important treatment modality of hematological malignancies, especially T-cell acute lymphoblastic leukemia (T-ALL). Glucocorticoids are known to induce a cell cycle arrest and apoptosis in T-lymphoma cells. We could demonstrate that the cell cycle arrest induced by the synthetic glucocorticoid dexamethasone (Dex) clearly precedes apoptosis in human CEM T-ALL and murine S49.1 T-lymphoma cells. Cyclin D3 is strongly downregulated, whereas the CDK inhibitor p27Kip1 (p27) is strongly upregulated in response to dexamethasone in these cells. RNAi-mediated knockdown of p27 as well as overexpression of its negative regulator Skp2 revealed the critical function of p27 in the Dex-induced G1 arrest of CEM cells. Our studies indicate that several mechanisms contribute to the increase of p27 protein in our T-lymphoma cell lines. We found a significant upregulation of p27 mRNA in S49.1 and CEM cells. In addition, Dex treatment activated the mouse p27 promotor in reporter gene experiments, indicating a transcriptional regulation. However, the relatively moderate induction of p27 mRNA levels by Dex did not explain the strong increase of p27 protein in CEM and S49.1 cells. We found clear evidence for a posttranslational mechanism responsible for the robust increase in p27 protein. Dex treatment of S49.1 and CEM cells increases the half-life of p27 protein, which indicates that decreased protein degradation is the primary mechanism of p27 induction by glucocorticoids. Interestingly, we found that Dex treatment decreased the protein and mRNA levels of the negative regulator of p27 protein and E3 ubiquitin ligase subunit Skp2. We conclude that the cell cycle inhibitor p27 and its negative regulator Skp2 are key players in the glucocorticoid-induced growth suppression of T-lymphoma cells and should be considered as potential drug targets to improve therapies of T-cell malignancies.
Collapse
Affiliation(s)
- Michael K Kullmann
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Karmakar S, Jin Y, Nagaich AK. Interaction of glucocorticoid receptor (GR) with estrogen receptor (ER) α and activator protein 1 (AP1) in dexamethasone-mediated interference of ERα activity. J Biol Chem 2013; 288:24020-34. [PMID: 23814048 PMCID: PMC3745347 DOI: 10.1074/jbc.m113.473819] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The role of glucocorticoids in the inhibition of estrogen (17-β-estradiol (E2))-regulated estrogen receptor (ER)-positive breast cancer cell proliferation is well established. We and others have seen that synthetic glucocorticoid dexamethasone (Dex) antagonizes E2-stimulated endogenous ERα target gene expression. However, how glucocorticoids negatively regulate the ERα signaling pathway is still poorly understood. ChIP studies using ERα- and glucocorticoid receptor (GR)-positive MCF-7 cells revealed that GR occupies several ERα-binding regions (EBRs) in cells treated with E2 and Dex simultaneously. Interestingly, there was little or no GR loading to these regions when cells were treated with E2 or Dex alone. The E2+Dex-dependent GR recruitment is associated with the displacement of ERα and steroid receptor coactivator-3 from the target EBRs leading to the repression of ERα-mediated transcriptional activation. The recruitment of GR to EBRs requires assistance from ERα and FOXA1 and is facilitated by AP1 binding within the EBRs. The GR binding to EBRs is mediated via direct protein-protein interaction between the GR DNA-binding domain and ERα. Limited mutational analyses indicate that arginine 488 located within the C-terminal zinc finger domain of the GR DNA-binding domain plays a critical role in stabilizing this interaction. Together, the results of this study unravel a novel mechanism involved in glucocorticoid inhibition of ERα transcriptional activity and E2-mediated cell proliferation and thus establish a foundation for future exploitation of the GR signaling pathway in the treatment of ER-positive breast cancer.
Collapse
Affiliation(s)
- Sudipan Karmakar
- Division of Therapeutic Proteins, Office of Biotechnology Products, Office of Pharmaceutical Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|
167
|
Duportets L, Maria A, Vitecek S, Gadenne C, Debernard S. Steroid hormone signaling is involved in the age-dependent behavioral response to sex pheromone in the adult male moth Agrotis ipsilon. Gen Comp Endocrinol 2013; 186:58-66. [PMID: 23474331 DOI: 10.1016/j.ygcen.2013.02.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
Abstract
In most animals, including insects, male reproduction depends on the detection and processing of female-produced sex pheromones. In the male moth, Agrotis ipsilon, both behavioral response and neuronal sensitivity in the primary olfactory center, the antennal lobe (AL), to female sex pheromone are age- and hormone-dependent. In many animal species, steroids are known to act at the brain level to modulate the responsiveness to sexually relevant chemical cues. We aimed to address the hypothesis that the steroidal system and in particular 20-hydroxyecdysone (20E), the main insect steroid hormone, might also be involved in this olfactory plasticity. Therefore, we first cloned the nuclear ecdysteroid receptor EcR (AipsEcR) and its partner Ultraspiracle (AipsUSP) of A. ipsilon, the expression of which increased concomitantly with age in ALs. Injection of 20E into young sexually immature males led to an increase in both responsiveness to sex pheromone and amount of AipsEcR and AipsUSP in their ALs. Conversely, the behavioral response decreased in older, sexually mature males after injection of cucurbitacin B (CurB), an antagonist of the 20E/EcR/USP complex. Also, the amount of AipsEcR and AipsUSP significantly declined after treatment with CurB. These results suggest that 20E is involved in the expression of sexual behavior via the EcR/USP signaling pathway, probably acting on central pheromone processing in A. ipsilon.
Collapse
Affiliation(s)
- Line Duportets
- UMR 1272, UPMC-INRA, Physiologie de l'Insecte: Signalisation et Communication, Université Paris VI, Bâtiment A, 7 quai Saint Bernard, 75005 Paris, France
| | | | | | | | | |
Collapse
|
168
|
Man XY, Li W, Chen JQ, Zhou J, Landeck L, Zhang KH, Mu Z, Li CM, Cai SQ, Zheng M. Impaired nuclear translocation of glucocorticoid receptors: novel findings from psoriatic epidermal keratinocytes. Cell Mol Life Sci 2013; 70:2205-20. [PMID: 23334186 PMCID: PMC11113139 DOI: 10.1007/s00018-012-1255-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 12/19/2012] [Accepted: 12/27/2012] [Indexed: 01/21/2023]
Abstract
Psoriasis is a chronic proliferative skin disease and is usually treated with topical glucocorticoids, which act through the glucocorticoid receptor (GR), a component of the physiological systems essential for immune responses, differentiation, and homeostasis. To investigate the possible role of GR in the pathogenesis of psoriasis, normal and psoriatic lesional skin were recruited. Firstly, the immunolocalization of GR in the skin and cultured epidermal keratinocytes were determined by immunofluorescence. In normal skin and cultured human epidermal keratinocytes, intracellular GR is localized in the nuclei, while in psoriatic skin and cultured keratinocytes, GR is in the cytoplasm. Next, we investigated possible factors associated with the cytoplasmic distribution. We found that VEGF and IFN-γ led to impaired nuclear translocation of GR through p53 and microtubule-inhibitor, vincristine, and inhibited nuclear uptake of GR in normal keratinocytes. In addition to dexamethasone, interleukin (IL)-13 was also able to transfer GR into nuclei of psoriatic keratinocytes. Furthermore, discontinuation of dexamethasone induced cytoplasmic retention of GR in normal keratinocytes. In contrast, energy depletion of normal epidermal keratinocytes did not change the nuclear distribution of GR. To confirm our findings in vivo, an imiquimod-induced psoriasis-like skin mouse model was included. IL-13 ameliorated (but vincristine exacerbated) the skin lesions on the mouse. Taken together, our findings define that impaired nuclear translocation of GR is associated with VEGF, IFN-γ, p53, and microtubule. Therapeutic strategies designed to accumulate GR in the nucleus, such as IL-13, may be beneficial for the therapy of psoriasis.
Collapse
Affiliation(s)
- Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Wei Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Jia-Qi Chen
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Jiong Zhou
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Lilla Landeck
- Department of Dermatology, University of Osnabrueck, Osnabrueck, Germany
| | - Kai-Hong Zhang
- Department of Dermatology, Affiliated Hospital, Taishan Medical College, Taishan, China
| | - Zhen Mu
- Department of Dermatology, Affiliated Hospital, Taishan Medical College, Taishan, China
| | - Chun-Ming Li
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Sui-Qing Cai
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Min Zheng
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| |
Collapse
|
169
|
Melville JM, Moss TJM. The immune consequences of preterm birth. Front Neurosci 2013; 7:79. [PMID: 23734091 PMCID: PMC3659282 DOI: 10.3389/fnins.2013.00079] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/02/2013] [Indexed: 01/24/2023] Open
Abstract
Preterm birth occurs in 11% of live births globally and accounts for 35% of all newborn deaths. Preterm newborns have immature immune systems, with reduced innate and adaptive immunity; their immune systems may be further compromised by various factors associated with preterm birth. The immune systems of preterm infants have a smaller pool of monocytes and neutrophils, impaired ability of these cells to kill pathogens, and lower production of cytokines which limits T cell activation and reduces the ability to fight bacteria and detect viruses in cells, compared to term infants. Intrauterine inflammation is a major contributor to preterm birth, and causes premature immune activation and cytokine production. This can induce immune tolerance leading to reduced newborn immune function. Intrauterine inflammation is associated with an increased risk of early-onset sepsis and likely has long-term adverse immune consequences. Requisite medical interventions further impact on immune development and function. Antenatal corticosteroid treatment to prevent newborn respiratory disease is routine but may be immunosuppressive, and has been associated with febrile responses, reductions in lymphocyte proliferation and cytokine production, and increased risk of infection. Invasive medical procedures result in an increased risk of late-onset sepsis. Respiratory support can cause chronic inflammatory lung disease associated with increased risk of long-term morbidity. Colonization of the infant by microorganisms at birth is a significant contributor to the establishment of the microbiome. Caesarean section affects infant colonization, potentially contributing to lifelong immune function and well-being. Several factors associated with preterm birth alter immune function. A better understanding of perinatal modification of the preterm immune system will allow for the refinement of care to minimize lifelong adverse immune consequences.
Collapse
Affiliation(s)
- Jacqueline M. Melville
- The Ritchie Centre, Monash Institute of Medical Research, Monash UniversityClayton, VIC, Australia
| | - Timothy J. M. Moss
- The Ritchie Centre, Monash Institute of Medical Research, Monash UniversityClayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash UniversityClayton, VIC, Australia
| |
Collapse
|
170
|
Cluning C, Ward BK, Rea SL, Arulpragasam A, Fuller PJ, Ratajczak T. The helix 1-3 loop in the glucocorticoid receptor LBD is a regulatory element for FKBP cochaperones. Mol Endocrinol 2013; 27:1020-35. [PMID: 23686112 DOI: 10.1210/me.2012-1023] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The heat-shock protein 90 (Hsp90) cochaperone FK506-binding protein 52 (FKBP52) upregulates, whereas FKBP51 inhibits, hormone binding and nuclear targeting of the glucocorticoid receptor (GR). Decreased cortisol sensitivity in the guinea pig is attributed to changes within the helix 1 to helix 3 (H1-H3) loop of the guinea pig GR (gpGR) ligand-binding domain. It has been proposed that this loop serves as a contact point for FKBP52 and/or FKBP51 with receptor. We examined the role of the H1-H3 loop in GR activation by FKBP52 using a Saccharomyces cerevisiae model. The activity of rat GR (rGR) containing the gpGR H1-H3 loop substitutions was still potentiated by FKBP52, confirming the loop is not involved in primary FKBP52 interactions. Additional assays also excluded a role for other intervening loops between ligand-binding domain helices in direct interactions with FKBP52 associated with enhanced receptor activity. Complementary studies in FKBP51-deficient mouse embryo fibroblasts and HEK293 cells demonstrated that substitution of the gpGR H1-H3 loop residues into rGR dramatically increased receptor repression by FKBP51 without enhancing receptor-FKBP51 interaction and did not alter recruitment of endogenous Hsp90 and the p23 cochaperone to receptor complexes. FKBP51 suppression of the mutated rGR did not require FKBP51 peptidylprolyl cis-trans isomerase activity and was not disrupted by mutation of the FK1 proline-rich loop thought to mediate reciprocal FKBP influences on receptor activity. We conclude that the gpGR-specific mutations within the H1-H3 loop confer global changes within the GR-Hsp90 complex that favor FKBP51 repression over FKBP52 potentiation, thus identifying the loop as an important target for GR regulation by the FKBP cochaperones.
Collapse
Affiliation(s)
- Carmel Cluning
- Laboratory for Molecular Endocrinology, Western Australian Institute forMedical Research and the UWA Centre for Medical Research, The University of Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
171
|
Li A, Hardy R, Stoner S, Tuckermann J, Seibel M, Zhou H. Deletion of mesenchymal glucocorticoid receptor attenuates embryonic lung development and abdominal wall closure. PLoS One 2013; 8:e63578. [PMID: 23696835 PMCID: PMC3656055 DOI: 10.1371/journal.pone.0063578] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/04/2013] [Indexed: 11/23/2022] Open
Abstract
As a member of the nuclear hormone receptor superfamily of ligand-activated transcription factors, the glucocorticoid receptor (GR) is essential for normal embryonic development. To date, the role of mesenchymal glucocorticoid signaling during development has not been fully elucidated. In the present study, we investigated the role of the GR during embryogenesis specifically in mesenchymal tissues. To this aim, we crossed GRflox mice with Dermo1-Cre mice to generate GR(Dermo1) mice, where the GR gene was deleted within mesenchymal cells. Compared to their wild type littermates, GR(Dermo1) mice displayed severe pulmonary atelectasis, defects in abdominal wall formation resulting in intestinal herniation, abnormal extracellular matrix synthesis in connective tissues and high postnatal lethality. Lungs of GR(Dermo1) mice failed to progress from the canalicular to saccular stage, as evidenced by the presence of immature air sacs, thickened interstitial mesenchyme and an underdeveloped vascular network between E17.5 and E18.5. Furthermore, myofibroblasts and vascular smooth muscle cells, although present in normal numbers in GR(Dermo1) animals, were characterized by significantly reduced elastin synthesis, whilst epithelial lining cells of the immature saccules were poorly differentiated. A marked reduction in normal elastin and collagen deposits were also observed in connective tissues adjacent to the umbilical hernia. This study demonstrates that eliminating the GR in cells of the mesenchymal lineage results in marked effects on interstitial fibroblast function, including a significant decrease in elastin synthesis. This results in lung atelectasis and postnatal lethality, as well as additional and hitherto unrecognized developmental defects in abdominal wall formation. In addition, altered glucocorticoid signaling in the mesenchyme attenuates normal lung epithelial differentiation.
Collapse
Affiliation(s)
- Aiqing Li
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Rowan Hardy
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
- Centre for Endocrinology, Diabetes and Metabolism, Institute of Biomedical Research, University of Birmingham, Birmingham, United Kingdom
| | - Shihani Stoner
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| | - Jan Tuckermann
- Institute of General Zoology and Endocrinology University of Ulm, Ulm, Germany
| | - Markus Seibel
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
- Dept of Endocrinology & Metabolism, Concord Hospital, Sydney, Australia
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
172
|
Bertucci PY, Nacht AS, Alló M, Rocha-Viegas L, Ballaré C, Soronellas D, Castellano G, Zaurin R, Kornblihtt AR, Beato M, Vicent GP, Pecci A. Progesterone receptor induces bcl-x expression through intragenic binding sites favoring RNA polymerase II elongation. Nucleic Acids Res 2013; 41:6072-86. [PMID: 23640331 PMCID: PMC3695497 DOI: 10.1093/nar/gkt327] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Steroid receptors were classically described for regulating transcription by binding to target gene promoters. However, genome-wide studies reveal that steroid receptors-binding sites are mainly located at intragenic regions. To determine the role of these sites, we examined the effect of progestins on the transcription of the bcl-x gene, where only intragenic progesterone receptor-binding sites (PRbs) were identified. We found that in response to hormone treatment, the PR is recruited to these sites along with two histone acetyltransferases CREB-binding protein (CBP) and GCN5, leading to an increase in histone H3 and H4 acetylation and to the binding of the SWI/SNF complex. Concomitant, a more relaxed chromatin was detected along bcl-x gene mainly in the regions surrounding the intragenic PRbs. PR also mediated the recruitment of the positive elongation factor pTEFb, favoring RNA polymerase II (Pol II) elongation activity. Together these events promoted the re-distribution of the active Pol II toward the 3′-end of the gene and a decrease in the ratio between proximal and distal transcription. These results suggest a novel mechanism by which PR regulates gene expression by facilitating the proper passage of the polymerase along hormone-dependent genes.
Collapse
|
173
|
Activation of mitogen- and stress-activated kinase 1 is required for proliferation of breast cancer cells in response to estrogens or progestins. Oncogene 2013; 33:1570-80. [PMID: 23604116 DOI: 10.1038/onc.2013.95] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/08/2013] [Accepted: 01/18/2013] [Indexed: 12/13/2022]
Abstract
Growth of breast cancers is often dependent on ovarian steroid hormones making the tumors responsive to antagonists of hormone receptors. However, eventually the tumors become hormone independent, raising the need to identify downstream targets for the inhibition of tumor growth. One possibility is to focus on the signaling mechanisms used by ovarian steroid hormones to induce breast cancer cell proliferation. Here we report that the mitogen- and stress-activated kinase 1 (MSK1) could be a potential druggable target. Using the breast cancer cell line T47D, we show that estrogens (E2) and progestins activate MSK1, which forms a complex with the corresponding hormone receptor. Inhibition of MSK1 activity with H89 or its depletion by MSK1 short hairpin RNAs (shRNAs) specifically abrogates cell proliferation in response to E2 or progestins without affecting serum-induced cell proliferation. MSK1 activity is required for the transition from the G1- to the S-phase of the cell cycle and inhibition of MSK1 compromises both estradiol- and progestin-dependent induction of cell cycle genes. ChIP-seq experiments identified binding of MSK1 to progesterone receptor-binding sites associated with hormone-responsive genes. MSK1 recruitment to epigenetically defined enhancer regions supports the need of MSK1 as a chromatin remodeler in hormone-dependent regulation of gene transcription. In agreement with this interpretation, expression of a histone H3 mutated at S10 eliminates the hormonal effect on cell proliferation and on induction of relevant target genes. Finally, we show that E2- or progestin-dependent growth of T47D cells xenografted in immunodefficient mice is inhibited by depletion of MSK1, indicating that our findings are not restricted to cultured cells, and that MSK1 plays an important role for hormone-dependent breast cancer growth in a more physiological context.
Collapse
|
174
|
Qin C, Lan X, He J, Xia X, Tian Y, Pei Z, Yuan H, Zhang Y. An in vitro and in vivo evaluation of a reporter gene/probe system hERL/(18)F-FES. PLoS One 2013; 8:e61911. [PMID: 23593502 PMCID: PMC3625158 DOI: 10.1371/journal.pone.0061911] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Accepted: 03/14/2013] [Indexed: 11/18/2022] Open
Abstract
Purpose To evaluate the feasibility of a reporter gene/probe system, namely the human estrogen receptor ligand binding domain (hERL)/16α-[18F] fluoro-17β-estradiol (18F-FES), for monitoring gene and cell therapy. Methods The recombinant adenovirus vector Ad5-hERL-IRES-VEGF (Ad-EIV), carrying a reporter gene (hERL) and a therapeutic gene (vascular endothelial growth factor, VEGF165) through an internal ribosome entry site (IRES), was constructed. After transfection of Ad-EIV into bone marrow mesenchymal stem cells (Ad-EIV-MSCs), hERL and VEGF165 mRNA and protein expressions were identified using Real-Time qRT-PCR and immunofluorescence. The uptake of 18F-FES was measured in both Ad-EIV-MSCs and nontransfected MSCs after different incubation time. Micro-PET/CT images were obtained at 1 day after injection of Ad-EIV-MSCs into the left foreleg of the rat. The right foreleg was injected with nontransfected MSCs, which served as self-control. Results After transfection with Ad-EIV, the mRNA and protein expression of hERL and VEGF165 were successfully detected in MSCs, and correlated well with each other (R2 = 0.9840, P<0.05). This indicated the reporter gene could reflect the therapeutic gene indirectly. Ad-EIV-MSCs uptake of 18F-FES increased with incubation time with a peak value of 9.13%±0.33% at 150 min, which was significantly higher than that of the control group. A far higher level of radioactivity could be seen in the left foreleg on the micro-PET/CT image than in the opposite foreleg. Conclusion These preliminary in vitro and in vivo studies confirmed that hERL/18F-FES might be used as a novel reporter gene/probe system for monitoring gene and cell therapy. This imaging platform may have broad applications for basic research and clinical studies.
Collapse
Affiliation(s)
- Chunxia Qin
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail:
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, School of Medicine, Charlottesville, Virginia, United States of America
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueli Tian
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijun Pei
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yuan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
175
|
Zhang Y, Leung DYM, Goleva E. Vitamin D enhances glucocorticoid action in human monocytes: involvement of granulocyte-macrophage colony-stimulating factor and mediator complex subunit 14. J Biol Chem 2013; 288:14544-14553. [PMID: 23572530 DOI: 10.1074/jbc.m112.427054] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Vitamin D (VitD) is now recognized for its pleiotrophic roles in regulating immune function. VitD interaction with other steroid receptor superfamily receptors in peripheral blood mononuclear cells is poorly understood. In the current study, we demonstrate that VitD enhanced glucocorticoid (GC) responses in human peripheral blood mononuclear cells because it stimulated GC induction of mitogen-activated protein kinase phosphatase-1 (MKP-1) and enhanced GC inhibition of LPS-induced IL-6. These VitD effects were abolished in purified CD14(+) and CD14(-) cells but were recovered in CD14(+) cells co-cultured with CD14(-) cells separated by tissue culture inserts. GM-CSF, found in culture supernatants from CD14(-) cells, was shown to mediate VitD enhancement of GC-induced MKP-1 production in monocytes via increased production of mediator complex subunit 14 (MED14). Recruitment of VitD receptor and MED14, 4.7 kbp upstream of the human MKP-1 gene transcription start site, enhanced binding of glucocorticoid receptor and histone H4 acetylation at the 4.6-kbp glucocorticoid response element of the MKP-1 promoter in the presence of GM-CSF in U937 cells. Knockdown of MED14 abolished VitD-mediated enhancement of GC-induced MKP-1 production. These data demonstrate VitD-mediated stimulation of GC anti-inflammatory effects in human monocytes and identify a role for GM-CSF and MED14 as mediators of this process.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206; Department of Pediatrics, University of Colorado Denver, Aurora, Colorado 80045
| | - Elena Goleva
- Department of Pediatrics, National Jewish Health, Denver, Colorado 80206.
| |
Collapse
|
176
|
Müller N, Fischer HJ, Tischner D, van den Brandt J, Reichardt HM. Glucocorticoids Induce Effector T Cell Depolarization via ERM Proteins, Thereby Impeding Migration and APC Conjugation. THE JOURNAL OF IMMUNOLOGY 2013; 190:4360-70. [DOI: 10.4049/jimmunol.1201520] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
177
|
Nishimura K, Nakano N, Chowdhury VS, Kaneto M, Torii M, Hattori MA, Yamauchi N, Kawai M. Effect of PPARβ/δ Agonist on the Placentation and Embryo-Fetal Development in Rats. ACTA ACUST UNITED AC 2013; 98:164-9. [DOI: 10.1002/bdrb.21052] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 01/08/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Kyohei Nishimura
- Drug Safely Evaluation; Drug Developmental Research Laboratories; Toyonaka; Osaka; Japan
| | - Nao Nakano
- Drug Safely Evaluation; Drug Developmental Research Laboratories; Toyonaka; Osaka; Japan
| | - Vishwajit Sur Chowdhury
- Department of Animal and Marine Bioresource Sciences; Graduate School Kyushu University; Hakozaki; Higashi-ku, Fukuoka; Japan
| | - Masako Kaneto
- Drug Safely Evaluation; Drug Developmental Research Laboratories; Toyonaka; Osaka; Japan
| | - Mikinori Torii
- Drug Safely Evaluation; Drug Developmental Research Laboratories; Toyonaka; Osaka; Japan
| | - Masa-aki Hattori
- Department of Animal and Marine Bioresource Sciences; Graduate School Kyushu University; Hakozaki; Higashi-ku, Fukuoka; Japan
| | - Nobuhiko Yamauchi
- Department of Animal and Marine Bioresource Sciences; Graduate School Kyushu University; Hakozaki; Higashi-ku, Fukuoka; Japan
| | - Motoyuki Kawai
- Drug Safely Evaluation; Drug Developmental Research Laboratories; Toyonaka; Osaka; Japan
| |
Collapse
|
178
|
Patki M, Chari V, Sivakumaran S, Gonit M, Trumbly R, Ratnam M. The ETS domain transcription factor ELK1 directs a critical component of growth signaling by the androgen receptor in prostate cancer cells. J Biol Chem 2013; 288:11047-65. [PMID: 23426362 DOI: 10.1074/jbc.m112.438473] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The androgen receptor (AR) is essential for diverse aspects of prostate development and function. Molecular mechanisms by which prostate cancer (PC) cells redirect AR signaling to genes that primarily support growth are unclear. A systematic search for critical AR-tethering proteins led to ELK1, an ETS transcription factor of the ternary complex factor subfamily. Although genetically redundant, ELK1 was obligatory for AR-dependent growth and clonogenic survival in both hormone-dependent PC and castration-recurrent PC cells but not for AR-negative cell growth. AR required ELK1 to up-regulate a major subset of its target genes that was strongly and primarily enriched for cell growth functions. AR functioned as a coactivator of ELK1 by association through its A/B domain, bypassing the classical mechanism of ELK1 activation by phosphorylation and without inducing ternary complex target genes. The ELK1-AR synergy per se was ligand-independent, although it required ligand for nuclear localization of AR as targeting the AR A/B domain to the nucleus recapitulated the action of hormone; accordingly, Casodex was a poor antagonist of the synergy. ELK3, the closest substitute for ELK1 in structure/function and genome recognition, did not interact with AR. ELK1 thus directs selective and sustained gene induction that is a substantial and critical component of growth signaling by AR in PC cells. The ELK1-AR interaction offers a functionally tumor-selective drug target.
Collapse
Affiliation(s)
- Mugdha Patki
- Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201-2013, USA
| | | | | | | | | | | |
Collapse
|
179
|
Chen CY, Tsai MM, Chi HC, Lin KH. Biological significance of a thyroid hormone-regulated secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2271-84. [PMID: 23429180 DOI: 10.1016/j.bbapap.2013.02.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 02/07/2013] [Accepted: 02/11/2013] [Indexed: 01/18/2023]
Abstract
The thyroid hormone, 3,3,5-triiodo-L-thyronine (T3), modulates several physiological processes, including cellular growth, differentiation, metabolism and proliferation, via interactions with thyroid hormone response elements (TREs) in the regulatory regions of target genes. Several intracellular and extracellular protein candidates are regulated by T3. Moreover, T3-regulated secreted proteins participate in physiological processes or cellular transformation. T3 has been employed as a marker in several disorders, such as cardiovascular disorder in chronic kidney disease, as well as diseases of the liver, immune system, endocrine hormone metabolism and coronary artery. Our group subsequently showed that T3 regulates several tumor-related secretory proteins, leading to cancer progression via alterations in extracellular matrix proteases and tumor-associated signaling pathways in hepatocellular carcinomas. Therefore, elucidation of T3/thyroid hormone receptor-regulated secretory proteins and their underlying mechanisms in cancers should facilitate the identification of novel therapeutic targets. This review provides a detailed summary on the known secretory proteins regulated by T3 and their physiological significance. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Department of Biochemistry, College of Medicine, Chang-Gung University, Taoyuan 333, Taiwan
| | | | | | | |
Collapse
|
180
|
Mechanistic basis and functional roles of long-term plasticity in auditory neurons induced by a brain-generated estrogen. J Neurosci 2013; 32:16478-95. [PMID: 23152630 DOI: 10.1523/jneurosci.3233-12.2012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The classic estrogen 17β-estradiol (E2) was recently identified as a novel modulator of hearing function. It is produced rapidly, in an experience-dependent fashion, by auditory cortical neurons of both males and females. This brain-generated E2 enhances the efficiency of auditory coding and improves the neural and behavioral discrimination of auditory cues. Remarkably, the effects of E2 are long-lasting and persist for hours after local rises in hormone levels have subsided. The mechanisms and functional consequences of this E2-induced plasticity of auditory responses are unknown. Here, we addressed these issues in the zebra finch model by combining intracerebral pharmacology, biochemical assays, in vivo neurophysiology in awake animals, and computational and information theoretical approaches. We show that auditory experience activates the MAPK pathway in an E2-dependent manner. This effect is mediated by estrogen receptor β (ERβ), which directly associates with MEKK1 to sequentially modulate MEK and ERK activation, where the latter is required for the engagement of downstream molecular targets. We further show that E2-mediated activation of the MAPK cascade is required for the long-lasting enhancement of auditory-evoked responses in the awake brain. Moreover, a functional consequence of this E2/MAPK activation is to sustain enhanced information handling and neural discrimination by auditory neurons for several hours following hormonal challenge. Our results demonstrate that brain-generated E2 engages, via a nongenomic interaction between an estrogen receptor and a kinase, a persistent form of experience-dependent plasticity that enhances the neural coding and discrimination of behaviorally relevant sensory signals in the adult vertebrate brain.
Collapse
|
181
|
Grzmil P, Altmann ME, Adham IM, Engel U, Jarry H, Schweyer S, Wolf S, Mänz J, Engel W. Embryo implantation failure and other reproductive defects in Ube2q1-deficient female mice. Reproduction 2013; 145:45-56. [PMID: 23108111 DOI: 10.1530/rep-12-0054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The ubiquitination process is indispensable for proteome regulation. Three classes of ubiquitin (Ub)-related proteins can be distinguished: E1, E2 and E3. Proteins from the E2 class are responsible for the transfer of Ubls from E1 to the target protein. For this activity, interaction with class E3 ligases is usually required. Ub-conjugating enzyme E2Q 1 (UBE2Q1) belongs to the E2 class of Ub-related enzymes and is demonstrated to be involved in the regulation of membrane B4GALT1 protein. Here, we demonstrate that human UBE2Q1 and mouse Ube2q1 are widely expressed and highly conserved genes. To elucidate the function of UBE2Q1 protein, we generated knockout mouse model. No overt phenotype was detected in UBE2Q1-deficient males, but in mutant females, pleiotropic reproductive defects were observed including altered oestrus cycle, abnormal sexual behaviour and reduced offspring care. Moreover, in the uterus of mutant females, significantly increased embryonic lethality and decreased implantation capacity of homozygous mutant embryos were noticed. We found that Ube2q1 is not expressed in the uterus of non-pregnant females but its expression is up-regulated during pregnancy. Taken together, Ube2q1 is involved in different aspects of female fertility.
Collapse
Affiliation(s)
- Pawel Grzmil
- Institute of Human Genetics, University of Göttingen, Heinrich Düker Weg 12, 37073 Göttingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Nixon M, Andrew R, Chapman KE. It takes two to tango: dimerisation of glucocorticoid receptor and its anti-inflammatory functions. Steroids 2013; 78:59-68. [PMID: 23127816 DOI: 10.1016/j.steroids.2012.09.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Revised: 08/28/2012] [Accepted: 09/07/2012] [Indexed: 01/30/2023]
Abstract
For a number of years, there has been a widespread view that the adverse side-effects of prolonged glucocorticoid (GC) treatment are a result of glucocorticoid receptor (GR)-mediated gene activation, whilst the beneficial anti-inflammatory effects result from GR-mediated 'transrepression'. Since the introduction of the dimerisation-deficient GR mutant, GR(dim), was apparently unable to activate gene transcription, yet still able to repress pro-inflammatory gene transcription, the search for novel GR modulators has centred on the separation of gene activation from repression by prevention of GR dimerisation. However, recent work has questioned the conclusions drawn from these early GR(dim) studies, with evidence that GR(dim) mutants not only activate gene transcription, but that, in direct contradiction to the initial GR(dim) work, are also capable of forming dimers. This review of the current literature highlights the versatility of the GR in forming homodimer interactions, as well as the ability to bind to alternate nuclear receptors, and investigates the potential implications such varying GR dimer conformations may have for the design of GR ligands with a safer side effect profile.
Collapse
Affiliation(s)
- Mark Nixon
- Endocrinology, University/British Heart Foundation Centre for Cardiovascular Science, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | | | | |
Collapse
|
183
|
Ballaré C, Castellano G, Gaveglia L, Althammer S, González-Vallinas J, Eyras E, Le Dily F, Zaurin R, Soronellas D, Vicent G, Beato M. Nucleosome-Driven Transcription Factor Binding and Gene Regulation. Mol Cell 2013. [DOI: 10.1016/j.molcel.2012.10.019] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
184
|
|
185
|
Vodo S, Arcelli D, Fiorenzani P, Meriggiola MC, Butkevich I, Di Canio C, Mikhailenko V, Aloisi AM. Gonadal ERα/β, AR and TRPV1 gene expression: modulation by pain and morphine treatment in male and female rats. Physiol Behav 2012; 110-111:80-6. [PMID: 23287630 DOI: 10.1016/j.physbeh.2012.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/27/2012] [Accepted: 12/13/2012] [Indexed: 12/14/2022]
Abstract
The results of several studies strongly indicate a bidirectional relationship among gonadal hormones and pain. While gonadal hormones play a key role in pain modulation, they have been found to be affected by pain therapies in different experimental and clinical conditions. However, the effects of pain and pain therapy on the gonads are still not clear. In this study, we determined the long-lasting (72 h) effects of inflammatory pain (formalin test) and/or morphine on estrogen receptor (ER), androgen receptor (AR) and TRPV1 gene expression in the rat testis and ovary. The animals were divided into groups: animals receiving no treatment, animals exposed only to the experimental procedure (control group), animals receiving no pain but morphine (sham/morphine), animals receiving pain and morphine (formalin/morphine), and animals receiving only formalin (formalin/saline). Testosterone (T) and estradiol (E) were determined in the plasma at the end of the testing. In the sham/morphine rats, there were increases of ERα, ERβ, AR and TRPV1 mRNA expression in the ovary; in the testis, ERα and ERβ mRNA expression were reduced while AR and TRPV1 expression were unaffected by treatment. T and E plasma levels were increased in morphine-treated female rats, while T levels were greatly reduced in morphine-treated and formalin-treated males. In conclusion, both testicular and ovarian ER (ERα and ERβ) and ovarian AR and TRPV1 gene expression appear to be affected by morphine treatment, suggesting long-lasting interactions among opioids and gonads.
Collapse
Affiliation(s)
- Stella Vodo
- Pain and Stress Neurophysiology Lab., Department of Physiology, University of Siena, 53100 Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Sivukhina E, Schäfer HH, Jirikowski GF. Differences in colocalization of corticosteroid-binding globulin and glucocorticoid receptor immunoreactivity in the rat brain. Ann Anat 2012; 195:219-24. [PMID: 23279724 DOI: 10.1016/j.aanat.2012.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 10/17/2012] [Accepted: 10/17/2012] [Indexed: 12/22/2022]
Abstract
Endocrine regulation of central and systemic stress response as well as learning and memory are in part controlled by systemic glucocorticoid levels. So far steroids have been thought to act on the brain predominantly through nuclear receptors. However, some brain systems known to respond to glucocorticoids seem to be devoid of the respective receptor proteins (GR). It is likely that known central actions of adrenal steroids may also be mediated by non-genomic actions involving intrinsic binding globulins. In recent studies we described the intrinsic expression of corticosteroid-binding globulin (CBG) in rat, mouse and human brains. Here we report an immunohistochemical mapping study on the colocalization of CBG and of GR in the rat brain. In the nucleus accumbens, septum, hippocampus, globus pallidus, medial and basolateral amygdale nuclei, magnocellular preoptic nuclei, diagonal band of Broca high intensity of CBG immunoreactivity was accompanied by weak or moderate GR staining, and vice versa. In the caudate putamen, bed nucleus of stria terminalis, septohypothalamic nucleus and parvocellular subdivision of the paraventricular nucleus strong GR immunoreactivity was observed, but CBG was almost undetectable. In contrast, throughout the supraoptic nucleus and magnocellular subdivision of the paraventricular nucleus numerous strongly CBG-positive cells were observed, devoid of specific GR immunoreactivity. It is most likely that CBG in the brain may be involved in the response to changing systemic glucocorticoid levels in addition to known nuclear and membrane corticosteroid receptors, or in glucocorticoid responsive regions devoid of these receptors.
Collapse
Affiliation(s)
- Elena Sivukhina
- Department of Anatomy II, Friedrich-Schiller University, Jena, Germany.
| | | | | |
Collapse
|
187
|
Hasan KMM, Rahman MS, Arif KMT, Sobhani ME. Psychological stress and aging: role of glucocorticoids (GCs). AGE (DORDRECHT, NETHERLANDS) 2012; 34:1421-1433. [PMID: 21971999 PMCID: PMC3528378 DOI: 10.1007/s11357-011-9319-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 09/19/2011] [Indexed: 05/31/2023]
Abstract
Psychological stress has extreme adverse consequences on health. However, the molecular mechanisms that mediate and accelerate the process of aging due to stress hormone are not well defined. This review has focused on diverse molecular paths that come out in response to chronic psychological stress via releasing of excessive glucocorticoids (GCs), involved in the aging process. GCs suppress transcription of nuclear cell adhesion molecules which impair synaptic plasticity, memory formation, and cognitive ability. Again, GCs promote muscle atrophy by means of motivating ubiquitin proteasome system and can repress muscle protein synthesis by inhibition of PI3-kinase/Akt pathway. GCs also inhibit interleukin-2 synthesis through suppressing T cell receptor signal that leads to loss of T cell activation, proliferation, and B-cell activation. Moreover, GCs increase the expression of collagenase-3, RANK ligand, and colony stimulating factor-1 that induce bone resorption. In general, stress-induced GCs can play causal role for aging and age-related disorders.
Collapse
Affiliation(s)
- K. M. Mehedi Hasan
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208 Bangladesh
| | - Md. Shaifur Rahman
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208 Bangladesh
| | - K. M. T. Arif
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208 Bangladesh
| | - Mahbub E. Sobhani
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, 9208 Bangladesh
| |
Collapse
|
188
|
Varayoud J, Monje L, Moreno-Piovano GS, Galoppo GH, Luque EH, Muñoz-de-Toro M, Ramos JG. Sexually dimorphic expression of receptor-alpha in the cerebral cortex of neonatal Caiman latirostris (Crocodylia: Alligatoridae). Gen Comp Endocrinol 2012; 179:205-13. [PMID: 22964531 DOI: 10.1016/j.ygcen.2012.08.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 08/01/2012] [Accepted: 08/20/2012] [Indexed: 01/30/2023]
Abstract
In mammals, estrogens have been described as endocrine and paracrine modulators of neuronal differentiation and synapse formation. However, the functional role of circulating estrogens and the distribution of estrogen receptors (ERs) in the cerebral cortex of reptiles have not been clearly established. Caiman latirostris (C. latirostris) is a South American species that presents temperature-dependent sex determination (TSD). By using immunohistochemistry, we have studied the distribution of ERα in the cerebral cortex of neonatal caimans. We studied brain samples from ten-day-old TSD-females and TSD-males and from female caimans that were administered estradiol during embryonic development (hormone-dependent sex determination, HSD-females). ERα was detected in the medial (MC), dorsal (DC) and lateral (LC) cortices. ERα expression in the MC showed sex-associated differences, being significantly greater in TSD-females compared to TSD-males. Interestingly, the highest ERα expression in the MC was exhibited by HSD-females. In addition, the circulating levels of estradiol were significantly higher in females (both TSD and HSD) than in TSD-males. Double immunostaining showed that ERα is expressed by neural precursor cells (as detected by ERα/doublecortin or ERα/glial fibrillary acidic protein) and mature neurons (ERα/neuron-specific nuclear protein). Our results demonstrate that the expression of ERα in the neonatal caiman cortex is sexually dimorphic and is present in the early stages of neuronal differentiation.
Collapse
Affiliation(s)
- Jorgelina Varayoud
- Laboratorio de Endocrinología y Tumores Hormonodependientes, School of Biochemistry and Biological Sciences, Universidad Nacional del Litoral, Santa Fe, Argentina
| | | | | | | | | | | | | |
Collapse
|
189
|
Abstract
Glucocorticoids have potent anti-inflammatory effects and have been used to treat patients with rheumatoid arthritis for more than 60 years. However, severe adverse effects of glucocorticoid treatment, including loss of bone mass and increased risk of fractures, are common. Data from studies of glucocorticoid-mediated gene regulation, which utilized conditional knockout mice in animal models of arthritis or glucocorticoid-induced osteoporosis, have substantially increased our understanding of the mechanisms by which glucocorticoids act via the glucocorticoid receptor. Following glucocorticoid binding, the receptor regulates gene expression either by interacting with DNA-bound transcription factors as a monomer or by binding directly to DNA as a dimer. In contrast to the old hypothesis that transrepression mechanisms involving monomeric glucocorticoid receptor actions were responsible for the anti-inflammatory effects of glucocorticoids, whereas dimeric glucocorticoid receptor binding resulted in adverse effects, data from animal models have shown that the anti-inflammatory and adverse effects of glucocorticoids are mediated by both monomeric and dimeric glucocorticoid receptor binding. This improved knowledge of the molecular mechanisms that underlie the beneficial and adverse effects of glucocorticoid therapy might lead to the development of rationales for novel glucocorticoid receptor ligands that could potentially have anti-inflammatory efficacy without adverse effects on bone.
Collapse
|
190
|
Sinchak K, Wagner EJ. Estradiol signaling in the regulation of reproduction and energy balance. Front Neuroendocrinol 2012; 33:342-63. [PMID: 22981653 PMCID: PMC3496056 DOI: 10.1016/j.yfrne.2012.08.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 08/18/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Our knowledge of membrane estrogenic signaling mechanisms and their interactions that regulate physiology and behavior has grown rapidly over the past three decades. The discovery of novel membrane estrogen receptors and their signaling mechanisms has started to reveal the complex timing and interactions of these various signaling mechanisms with classical genomic steroid actions within the nervous system to regulate physiology and behavior. The activation of the various estrogenic signaling mechanisms is site specific and differs across the estrous cycle acting through both classical genomic mechanisms and rapid membrane-initiated signaling to coordinate reproductive behavior and physiology. This review focuses on our current understanding of estrogenic signaling mechanisms to promote: (1) sexual receptivity within the arcuate nucleus of the hypothalamus, (2) estrogen positive feedback that stimulates de novo neuroprogesterone synthesis to trigger the luteinizing hormone surge important for ovulation and estrous cyclicity, and (3) alterations in energy balance.
Collapse
Affiliation(s)
- Kevin Sinchak
- Department of Biological Sciences, California State University, Long Beach, 1250 Bellflower Blvd., Long Beach, CA 90840-9502, United States.
| | | |
Collapse
|
191
|
Dual-color bioluminescent bioreporter for forensic analysis: evidence of androgenic and anti-androgenic activity of illicit drugs. Anal Bioanal Chem 2012; 405:1035-45. [DOI: 10.1007/s00216-012-6416-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 09/06/2012] [Accepted: 09/06/2012] [Indexed: 12/19/2022]
|
192
|
Cruz SA, Chao PL, Hwang PP. Cortisol promotes differentiation of epidermal ionocytes through Foxi3 transcription factors in zebrafish (Danio rerio). Comp Biochem Physiol A Mol Integr Physiol 2012; 164:249-57. [PMID: 23010242 DOI: 10.1016/j.cbpa.2012.09.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2012] [Revised: 09/14/2012] [Accepted: 09/17/2012] [Indexed: 01/30/2023]
Abstract
Glucocorticoid regulates epidermal cell proliferation, and is used to treat certain skin disorders. Cortisol, a glucocorticoid, is also linked to skin development in teleost fish. Cortisol increases the number of epithelial ionocytes during environmental acclimation in euryhaline fishes, but it is unclear whether this is due to increased differentiation or proliferation. To investigate, we treated zebrafish embryos with exogenous cortisol (20mg/L). The densities of the ionocytes Na(+)-K(+)-ATPase rich cells (NaRCs) and H(+)-ATPase rich cells (HRCs) were significantly increased by cortisol, and this was accompanied by an increase in the respective marker genes. Expression of the glucocorticoid receptor (GR) gene was decreased. Cortisol treatment also increased ionocytes in cultured adult zebrafish gills, and up-regulated expression of genes encoding forkhead box I3 (foxi3a and foxi3b) transcription factors, which regulate ionocyte progenitor development. GR expression was up-regulated by cortisol in vitro; as such, the observed decrease in vivo reflects a regulatory systemic-negative feedback. Notably, in situ hybridization revealed that foxi3a/b mRNA expression was increased by cortisol at 24-48h post-fertilization. Cortisol also decreased keratinocytes, but did not affect epidermal stem cells or mucus cells. We conclude that foxi3a/b transactivation by cortisol-GR favors differentiation of ionocyte progenitors, thereby facilitating proliferation of mature ionocytes.
Collapse
Affiliation(s)
- Shelly Abad Cruz
- Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei, Taiwan, ROC
| | | | | |
Collapse
|
193
|
Shah K, Patel D, Jadav P, Sheikh M, Sairam KVVM, Joharapurkar A, Jain MR, Bahekar R. Discovery of liver selective non-steroidal glucocorticoid receptor antagonist as novel antidiabetic agents. Bioorg Med Chem Lett 2012; 22:5857-62. [PMID: 22917520 DOI: 10.1016/j.bmcl.2012.07.078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 07/17/2012] [Accepted: 07/24/2012] [Indexed: 11/16/2022]
Affiliation(s)
- Kiran Shah
- Zydus Research Centre, Sarkhej-Bavla N.H. 8A Moraiya, Ahmedabad 382210, India
| | | | | | | | | | | | | | | |
Collapse
|
194
|
Khurana S, Chakraborty S, Zhao X, Liu Y, Guan D, Lam M, Huang W, Yang S, Kao HY. Identification of a novel LXXLL motif in α-actinin 4-spliced isoform that is critical for its interaction with estrogen receptor α and co-activators. J Biol Chem 2012; 287:35418-35429. [PMID: 22908231 DOI: 10.1074/jbc.m112.401364] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
α-Actinins (ACTNs) are a family of proteins cross-linking actin filaments that maintain cytoskeletal organization and cell motility. Recently, it has also become clear that ACTN4 can function in the nucleus. In this report, we found that ACTN4 (full length) and its spliced isoform ACTN4 (Iso) possess an unusual LXXLL nuclear receptor interacting motif. Both ACTN4 (full length) and ACTN4 (Iso) potentiate basal transcription activity and directly interact with estrogen receptor α, although ACTN4 (Iso) binds ERα more strongly. We have also found that both ACTN4 (full length) and ACTN4 (Iso) interact with the ligand-independent and the ligand-dependent activation domains of estrogen receptor α. Although ACTN4 (Iso) interacts efficiently with transcriptional co-activators such as p300/CBP-associated factor (PCAF) and steroid receptor co-activator 1 (SRC-1), the full length ACTN4 protein either does not or does so weakly. More importantly, the flanking sequences of the LXXLL motif are important not only for interacting with nuclear receptors but also for the association with co-activators. Taken together, we have identified a novel extended LXXLL motif that is critical for interactions with both receptors and co-activators. This motif functions more efficiently in a spliced isoform of ACTN4 than it does in the full-length protein.
Collapse
Affiliation(s)
- Simran Khurana
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Sharmistha Chakraborty
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Xuan Zhao
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Yu Liu
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Dongyin Guan
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Minh Lam
- Comprehensive Cancer Center, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Wei Huang
- Center for Proteomics and Department of Pharmacology, School of Medicine, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Sichun Yang
- Center for Proteomics and Department of Pharmacology, School of Medicine, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Hung-Ying Kao
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106.
| |
Collapse
|
195
|
Davis JR, Mossalam M, Lim CS. Utilizing the estrogen receptor ligand-binding domain for controlled protein translocation to the insoluble fraction. Pharm Res 2012; 29:3455-63. [PMID: 22869106 DOI: 10.1007/s11095-012-0840-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Accepted: 07/16/2012] [Indexed: 01/23/2023]
Abstract
PURPOSE The estrogen receptor forms insoluble aggregates in the insoluble cytoskeletal subcellular fraction when bound to the antagonist fulvestrant. The ligand-binding domain was isolated and fused to signal sequences to target subcellular compartments. Sequestering a pro-apoptotic peptide tested the utility of a protein targeted to the insoluble fraction. METHODS The ligand-binding domain of the estrogen receptor was isolated and fused with signal sequences, either a nuclear localization signal or nuclear export signal. The subcellular localization when bound to drug fulvestrant was examined, specifically its interaction with cytokeratins 8 and 18. The ability to target a therapeutic peptide to the insoluble fraction was tested by fusing a therapeutic coiled-coil from Bcr-Abl in K562 cells. RESULTS The estrogen receptor ligand-binding domain responds to fulvestrant by translocating to the insoluble fraction. Adding a signal sequence significantly limited the translocation to either the nucleus or cytoplasm. The cytokeratin 8/18 status of the cell did not alter this response. The therapeutic coiled-coil fused to ERLBD was inactivated upon ligand induction. CONCLUSIONS Isolating the ligand-binding domain of the estrogen receptor creates a ligand-controllable protein capable of translocation to the insoluble fraction. This can be used to sequester an active peptide to alter its function.
Collapse
Affiliation(s)
- James R Davis
- Department of Pharmaceutics and Pharmaceutical Chemistry College of Pharmacy, University of Utah, Salt Lake City, Utah, USA.
| | | | | |
Collapse
|
196
|
Src kinase-mediates androgen receptor-dependent non-genomic activation of signaling cascade leading to endothelial nitric oxide synthase. Biochem Biophys Res Commun 2012; 424:538-43. [PMID: 22771325 DOI: 10.1016/j.bbrc.2012.06.151] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 06/28/2012] [Indexed: 11/21/2022]
Abstract
Our previous study has demonstrated that testosterone rapidly activates endothelial nitric oxide synthase (eNOS), enhancing nitric oxide (NO) release from endothelial cells (ECs) via the phosphatidylinositol 3-kinase/Akt (PI3-kinase/Akt) pathway. The upstream regulators of this pathway are unknown. In this study, we further investigated the non-genomic action of testosterone in human aortic ECs. Acute (30 min) activation of eNOS caused by testosterone was unaffected by pretreatment with a transcriptional inhibitor, actinomycin D. Non-permeable testosterone-BSA rapidly induced Akt and eNOS phosphorylation. In contrast, luciferase reporter assay showed that the transcriptional activity of the androgen-responsive element (ARE) was increased by testosterone, but not by testosterone-BSA at 2h after stimulation. Immunostaining displayed co-localization of androgen receptor (AR) with caveolin-1. Fractional analysis showed that AR was expressed in caveolae-enriched membrane fractions. Immunoprecipitation assays revealed the association of AR with caveolin-1 and c-Src, suggesting complex formation among them. Testosterone rapidly increased the phosphorylation of c-Src on Tyr416, which was inhibited by an AR antagonist and by siRNA for AR. PP2, a specific-inhibitor of Src kinase, abolished the testosterone-induced phosphorylation of Akt and eNOS. Our data indicate that testosterone induces rapid assembly of a membrane signaling complex among AR, caveolin-1 and c-Src, which then facilitates activation of the c-Src/ PI3-kinase/Akt cascade, resulting in activation of eNOS.
Collapse
|
197
|
Abstract
The effects of glucocorticoid on lipid metabolism of broiler chicken (Gallus gallus domesticus) skeletal muscle were investigated. Male Arbor Acres chickens (35 days old) were subjected to dexamethasone treatment for 3 days. We found that dexamethasone retards body growth while facilitating lipid accumulation. In M. pectoralis major (PM), dexamethasone increased the expression of glucocorticoid receptor (GR), fatty acid transport protein 1 (FATP1), heart fatty acid-binding protein (H-FABP) and long-chain acyl-CoA dehydrogenase (LCAD) mRNA and decreased the expression of liver carnitine palmitoyltransferase 1 (L-CPT1), adenosine-monophosphate-activated protein kinase (AMPK) α2 and lipoprotein lipase (LPL) mRNA. LPL activity was also decreased. In M. biceps femoris (BF), the levels of GR, FATP1 and L-CPT1 mRNA were increased. AMPKα (Thr172) phosphorylation and CTP1 activity of skeletal muscle were decreased by dexamethasone. In fed chickens, dexamethasone enhanced very low-density lipoprotein receptor (VLDLR) expression and AMPK activity in muscle, but it impaired the expression of LPL and L-CPT1 mRNA and LPL activity in PM and augmented the expression of GR, LPL, H-FABP, L-CPT1, LCAD and AMPKα2 mRNA in BF. Adipose triglyceride lipase (ATGL) protein expression was not affected by dexamethasone. In conclusion, in the fasting state, dexamethasone-induced-retarded fatty acid utilisation may be involved in the augmented intramyocellular lipid accumulation in both glycolytic (PM) and oxidative (BF) muscle tissues. In the fed state, dexamethasone promoted the transcriptional activity of genes related to lipid uptake and oxidation in muscles. Unmatched lipid uptake and utilisation are suggested to be involved in the augmented intramyocellular lipid accumulation.
Collapse
Affiliation(s)
- X J Wang
- Department of Animal Science, Shandong Agricultural University, Taian, Shandong 271018, PR China
| | | | | | | |
Collapse
|
198
|
In silico design of peptidic inhibitors targeting estrogen receptor alpha dimer interface. Mol Divers 2012; 16:441-51. [PMID: 22752657 DOI: 10.1007/s11030-012-9378-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/28/2012] [Indexed: 10/28/2022]
Abstract
Human estrogen receptor alpha (ERα), which acts as a biomarker and as a therapeutic target for breast cancers, is activated by agonist ligands and co-activator proteins. Selective estrogen receptor modulators (SERM) act as antagonists in specific tissues and tamoxifen, a SERM, has served as a drug for decades for ERα-positive breast cancers. However, the ligand-selective and tissue-specific response of ERα biological activity and the resistance to tamoxifen treatment in advanced stages of ERα-positive breast cancers underscores the need to find a ligand-independent inhibitor for ERα. Here we present a ligand-independent approach of inhibiting ERα transactivation targeting its dimerization-a key process of ERα biological activity. Using in silico techniques, we first elucidated the hydrogen bond interactions involved in dimerization and identified three interfacial sequence motifs, where sequence I (DKITD) and sequence II (QQQHQRLAQ) of one monomer form hydrogen bonding with sequence II and sequence I of the second monomer, respectively, and sequence III (LSHIRHMSNK) hydrogen bonds with the same from the second monomer. Studying the structural stability and the binding affinity of the peptides derived from these sequence motifs, we found that an extended and ARG mutated version (LQQQHQQLAQ) of sequence II can act as a suitable template for designing peptidic inhibitors. It provides additional structural stability and interacts more strongly with ERα dimer interface groove formed by helices 9 and 10/11 and prevent ERα dimerization. Our result provides a novel therapeutic designing pipeline for ligand-independent inhibition of ERα.
Collapse
|
199
|
Glutamine synthetase activity and the expression of three glul paralogues in zebrafish during transport. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:274-84. [PMID: 22750401 DOI: 10.1016/j.cbpb.2012.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/18/2012] [Accepted: 06/19/2012] [Indexed: 01/13/2023]
Abstract
The enzyme glutamine synthetase (GS; glutamate-ammonia ligase, EC 6.3.1.2) plays an important role in the nitrogen metabolism of fish. In this study the GS activity and the corresponding genes were examined to understand how they are regulated in zebrafish in response to hyperammonemic stress during a 72 h simulated transport. Whole body ammonia levels, the activity of the enzyme GS and the mRNA expression of the splice variants of three paralogues of glul, glutamine synthetase gene (glula, glulb and glulc) were examined in brain, liver and kidney of zebrafish. Whole body ammonia reached significantly higher levels by 48 h, while brain showed higher levels as early as 24 h, compared to the values at the start of the transport. The GS activities in brain, liver and kidney were significantly higher at the end of 72 h transport than those at the start. However, only the expression of mRNA of glulb-002 and glulb-003 were significantly upregulated during the simulated transport. In silico analysis of the putative promoter regions of glul paralogues revealed glucocorticoid receptor binding sites. However, glucocorticoid response elements of glulb were not different. The up-regulation of GS enzyme activity and hitherto unreported mRNA expression of glul paralogues during zebrafish transport indicate a physiological response of fish to ammonia.
Collapse
|
200
|
Novella S, Dantas AP, Segarra G, Medina P, Hermenegildo C. Vascular Aging in Women: is Estrogen the Fountain of Youth? Front Physiol 2012; 3:165. [PMID: 22685434 PMCID: PMC3368545 DOI: 10.3389/fphys.2012.00165] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 05/08/2012] [Indexed: 11/13/2022] Open
Abstract
Aging is associated with structural and functional changes in the vasculature, including endothelial dysfunction, arterial stiffening and remodeling, impaired angiogenesis, and defective vascular repair, and with increased prevalence of atherosclerosis. Cardiovascular risk is similar for older men and women, but lower in women during their fertile years. This age- and sex-related difference points to estrogen as a protective factor because menopause is marked by the loss of endogenous estrogen production. Experimental and some clinical studies have attributed most of the protective effects of estrogen to its modulatory action on vascular endothelium. Estrogen promotes endothelial-derived NO production through increased expression and activity of endothelial nitric oxide synthase, and modulates prostacyclin and thromboxane A(2) release. The thromboxane A(2) pathway is key to regulating vascular tone in females. Despite all the experimental evidence, some clinical trials have reported no cardiovascular benefit from estrogen replacement therapy in older postmenopausal women. The "Timing Hypothesis," which states that estrogen-mediated vascular benefits occur only before the detrimental effects of aging are established in the vasculature, offers a possible explanation for these discrepancies. Nevertheless, a gap remains in current knowledge of cardiovascular aging mechanisms in women. This review comprises clinical and experimental data on the effects of aging, estrogens, and hormone replacement therapy on vascular function of females. We aim to clarify how menopause and aging contribute jointly to vascular aging and how estrogen modulates vascular response at different ages.
Collapse
Affiliation(s)
- Susana Novella
- Departamento de Fisiología, Universitat de València Valencia, Spain
| | | | | | | | | |
Collapse
|