2,148 results on '"Hormone response element"'
Search Results
2. The Estrogen-Responsive Transcriptome of Female Secondary Sexual Traits in the Gulf Pipefish.
- Author
-
Anderson, Andrew P, Rose, Emily, Flanagan, Sarah P, and Jones, Adam G
- Subjects
- *
BINDING site assay , *SEXUAL dimorphism , *MUSCLE growth , *ADIPOSE tissues , *CHROMATOPHORES , *GENE expression - Abstract
Sexual dimorphism often results from hormonally regulated trait differences between the sexes. In sex-role-reversed vertebrates, females often have ornaments used in mating competition that are expected to be under hormonal control. Males of the sex-role-reversed Gulf pipefish (Syngnathus scovelli) develop female-typical traits when they are exposed to estrogens. We aimed to identify genes whose expression levels changed during the development and maintenance of female-specific ornaments. We performed RNA-sequencing on skin and muscle tissue in male Gulf pipefish with and without exposure to estrogen to investigate the transcriptome of the sexually dimorphic ornament of vertical iridescent bands found in females and estrogen-exposed males. We further compared differential gene expression patterns between males and females to generate a list of genes putatively involved in the female secondary sex traits of bands and body depth. A detailed analysis of estrogen-receptor binding sites demonstrates that estrogen-regulated genes tend to have nearby cis-regulatory elements. Our results identified a number of genes that differed between the sexes and confirmed that many of these were estrogen-responsive. These estrogen-regulated genes may be involved in the arrangement of chromatophores for color patterning, as well as in the growth of muscles to achieve the greater body depth typical of females in this species. In addition, anaerobic respiration and adipose tissue could be involved in the rigors of female courtship and mating competition. Overall, this study generates a number of interesting hypotheses regarding the genetic basis of a female ornament in a sex-role-reversed pipefish. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
3. Introduction to Pediatric Endocrinology: The Endocrine System
- Author
-
Styne, Dennis M. and Styne, Dennis M.
- Published
- 2016
- Full Text
- View/download PDF
4. Sperm associated antigen 7 is activated by T3 during Xenopus tropicalis metamorphosis via a thyroid hormone response element within the first intron
- Author
-
LaTaijah Crawford, Nga Luu, Liezhen Fu, Andrew Tong, Yuta Tanizaki, and Yun-Bo Shi
- Subjects
Male ,Thyroid Hormones ,Xenopus ,media_common.quotation_subject ,Organogenesis ,Response Elements ,Article ,Xenopus laevis ,Transcription (biology) ,Animals ,Metamorphosis ,Receptor ,media_common ,Hormone response element ,Thyroid hormone receptor ,biology ,Metamorphosis, Biological ,Intron ,Gene Expression Regulation, Developmental ,Cell Biology ,biology.organism_classification ,Spermatozoa ,Introns ,Cell biology ,Triiodothyronine ,Developmental Biology - Abstract
Thyroid hormone (T3) affects many diverse physiological processes such as metabolism, organogenesis, and growth. The two highly related frog species, diploid Xenopus tropicalis and pseudo tetraploid Xenopus laevis, have been used as models for analyzing the effects of T3 during vertebrate development. T3 regulates T3-inducible gene transcription through T3 receptor (TR)-binding to T3-response elements (TREs). We have previously identified sperm associated antigen 7 (spag7) as a candidate T3 target gene that is potentially involved in adult stem cell development and/or proliferation during intestinal metamorphosis. To investigate whether T3 regulates spag7 directly at the transcriptional level via TR, we first conducted qRT-PCR to analyze its expression during natural and T3-induced metamorphosis and found that spag7 was up-regulated during natural metamorphosis in the intestine, tail, brain and hindlimb, peaking at the climax of metamorphosis in all those organs, and upon T3 treatment of premetamorphic tadpoles. Next, we demonstrated that an intronic TRE in spag7, first identified through bioinformatic analysis, could bind to TR in vitro and in vivo during metamorphosis. A dual luciferase assay utilizing a reconstituted frog oocyte transcription system showed that the TRE could mediate promoter activation by liganded TR. These results indicate that spag7 expression is directly regulated by T3 through the TRE in the first intron during metamorphosis, implicating a role for spag7 early during T3-regulated tissue remodeling and resorption.
- Published
- 2022
5. Regulation of Gene Expression
- Author
-
Chedrese, Pedro J. and Chedrese, Pedro J., editor
- Published
- 2009
- Full Text
- View/download PDF
6. A Profile HMM for Recognition of Hormone Response Elements
- Author
-
Stepanova, Maria, Lin, Feng, Lin, Valerie C. -L., Hutchison, David, editor, Kanade, Takeo, editor, Kittler, Josef, editor, Kleinberg, Jon M., editor, Mattern, Friedemann, editor, Mitchell, John C., editor, Naor, Moni, editor, Nierstrasz, Oscar, editor, Pandu Rangan, C., editor, Steffen, Bernhard, editor, Sudan, Madhu, editor, Terzopoulos, Demetri, editor, Tygar, Dough, editor, Vardi, Moshe Y., editor, Weikum, Gerhard, editor, Istrail, Sorin, editor, Pevzner, Pavel, editor, Waterman, Michael, editor, Rajapakse, Jagath C., editor, Wong, Limsoon, editor, and Acharya, Raj, editor
- Published
- 2006
- Full Text
- View/download PDF
7. Novel Compounds Acting Through Receptors and Enzyme Mediators
- Author
-
Penning, Trevor M., Lobo, R. A., editor, Crosignani, P. G., editor, Paoletti, R., editor, and Bruschi, F., editor
- Published
- 2002
- Full Text
- View/download PDF
8. Effects of neonatal dexamethasone and CpdA on the expression of genes for apoptosis regulator proteins in the neonatal hippocampus
- Author
-
Timofey A. Lagunov, E. V. Sukhareva, Dmitriy A. Lanshakov, Tatyana S. Kalinina, and V. V. Bulygina
- Subjects
Hormone response element ,medicine.medical_specialty ,Programmed cell death ,hippocampus ,Physiology ,Chemistry ,brain cell type markers ,Endocrinology ,Mineralocorticoid receptor ,Glucocorticoid receptor ,Apoptosis ,apoptosis regulator protein ,Internal medicine ,glucocorticoid receptor ,neocortex ,medicine ,QP1-981 ,Receptor ,development ,Transcription factor ,hormones, hormone substitutes, and hormone antagonists ,Transrepression - Abstract
Glucocorticoids (GC) are crucial regulators of homeostasis and function. Despite its negative side effects, glucocorticoid therapy in neonates is widely used antenatally for accelerating fetal lung maturation in cases of preterm birth. GC action is mediated via glucocorticoid receptors — ligand-activated transcription factors. Cell death and viability in the neonatal brain are regulated by many factors, but the glucocorticoid receptor signalling is high above them. The present work studies the changes in the expression of genes for apoptosis regulators with Bcl-2 homology (BH) domains (Bcl-xL, Bax, Bim, Bok, Bid) in the neonatal rat hippocampus after dexamethasone (DEX) and CpdA administration. CpdA is a dissociative ligand — glucocorticoid receptor modulator — that shifts glucocorticoid receptor (GR) activity toward transrepression. Ligands administration to P2 pups caused different patterns of timeline changes in the expression of the studied genes. We observed the first increase in the mRNA level of the genes which have glucocorticoid response element (GRE) (Bcl-xL, Bim) in their promoter 30 min after DEX administration. Activated GR action on cells in the neonatal hippocampus is complex and long-lasting; it could also contain receptor homo- and hetero-dimerisation. Using rat pheochromocytoma PC12 cells as a test system, we assessed GR-GR and GR-MR (mineralocorticoid receptor) dimerisation with proximity ligation assay (PLA) assay separately in the nucleus and cytoplasm after DEX and CpdA administration. An increase in GR-GR dimers in the cell nucleus was observed only after DEX administration. In the cell cytoplasm, we observed a gradual (DEX more than CpdA) increase in the number of both GR-GR and GR-MR dimers.
- Published
- 2021
9. Glucocorticoid receptor-mediated alleviation of inflammation by berberine: in vitro, in silico and in vivo investigations
- Author
-
Jie Zhang, Fangyu Li, Yuan Liang, Jingqi Zhao, Chenfei Li, Haoyang Zou, Tiehua Zhang, and Li Ren
- Subjects
Hormone response element ,Transactivation ,chemistry.chemical_compound ,Berberine ,Glucocorticoid receptor ,Chemistry ,In vivo ,In silico ,General Medicine ,In vitro ,Food Science ,Transrepression ,Cell biology - Abstract
As a natural dietary ingredient, berberine possesses multiple biological activities including anti-inflammatory effects. In this work, glucocorticoid receptor (GR)-mediated alleviation of inflammation by berberine was investigated by a combination of in vitro, in silico, and in vivo approaches. The fluorescence polarization assay showed that berberine bound to GR with an IC50 value of 9.14 ± 0.16 pM. Molecular docking and molecular dynamics simulation suggested that berberine bound stably to the active site of GR via hydrogen bonding and hydrophobic interactions. Berberine induced GR nuclear translocation but did not activate the glucocorticoid response element in HeLa cells. Furthermore, both gene and protein expressions of PEPCK were significantly attenuated by berberine in HepG2 cells. Interestingly, berberine downregulated CBG mRNA and protein levels without up-regulating TAT mRNA and protein levels in HepG2 cells, demonstrating its dissociated characteristics that could separate transrepression from transactivation. In addition, the in vitro and in vivo anti-inflammatory effects of berberine were confirmed in lipopolysaccharide-induced RAW 264.7 cells and in a mouse model of allergic contact dermatitis, respectively. In conclusion, berberine might serve as a potential selective GR modulator.
- Published
- 2021
10. LncRNA GAS5 is upregulated in polycystic ovary syndrome and regulates cell apoptosis and the expression of IL-6
- Author
-
Chunxia Wang, Yaru Jiang, Xiaoqian Zhang, Shishi Yue, Dongxu Pei, Zhijing Zhao, Yongwei Li, Xinwei Liu, and Yanjia Mao
- Subjects
0301 basic medicine ,Adult ,endocrine system diseases ,Cell ,Apoptosis ,Transfection ,lcsh:Gynecology and obstetrics ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Downregulation and upregulation ,GAS5 ,medicine ,Gene silencing ,Humans ,Polycystic ovary syndrome ,lcsh:RG1-991 ,Hormone response element ,IL-6 ,business.industry ,Interleukin-6 ,Research ,Obstetrics and Gynecology ,Granulosa ,Polycystic ovary ,Up-Regulation ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Cancer research ,Female ,RNA, Long Noncoding ,business ,Hormone - Abstract
Background GAS5 contains a hormone response element that can induce cell apoptosis in breast cancer. It is known that cell apoptosis and hormone response play crucial roles in polycystic ovary syndrome (PCOS), indicating the potential involvement of GAS5 in PCOS. This study was performed to investigate the potential involvement of GAS5 and IL-6 (a critical player in PCOS) in PCOS. Methods Research subjects of this study included 60 PCOS patients and 60 healthy controls. The expression levels of GAS5 and IL-6 in plasma of both patients and controls were measured by qPCR and ELISA, respectively. Cell transfections were performed to analyze the interaction between GAS5 and IL-6. Cell apoptosis was analyzed by cell apoptosis assay. Results GAS5 was upregulated in plasma of PCOS patients. The expression levels of GAS5 were positively correlated with the expression levels of IL-6. Altered expression levels of GAS5 and IL-6 distinguished PCOS patients from healthy controls. In cells of a granulosa-like tumor cell line (KGN), overexpression of GAS5 led to upregulated IL-6, while silencing of GAS5 played an opposite role. Cell apoptosis analysis showed that overexpression of GAS5 significantly decreased apoptosis rate of KGN cells. Silencing of GAS5 increased the rate of KGN cell apoptosis. Conclusions GAS5 is upregulated in PCOS and regulates cell apoptosis and the expression of IL-6.
- Published
- 2020
11. Neuroendocrine Regulation of Sexual Behavior
- Author
-
Pfaff, Donald W., Vasudevan, Nandini, Attardi, Barbara, Conn, P. Michael, editor, and Freeman, Marc E., editor
- Published
- 2000
- Full Text
- View/download PDF
12. Structure and Function of the Estrogen Receptor
- Author
-
Hihi, A. K., Wahli, W., Oettel, Michael, editor, and Schillinger, Ekkehard, editor
- Published
- 1999
- Full Text
- View/download PDF
13. SIRT2 suppresses expression of inflammatory factors via Hsp90‐glucocorticoid receptor signalling
- Author
-
Adil J. Nazarali, Yao lin, Xuan Wang, Ao Xu, Kai Sun, Xiaofang Zhao, Shaoping Ji, and Na Fang
- Subjects
0301 basic medicine ,Lipopolysaccharides ,inflammatory cytokine ,Lipopolysaccharide ,deacetylation ,Inflammation ,Hsp90 ,Glucocorticoid receptor ,SIRT2 ,Models, Biological ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Receptors, Glucocorticoid ,Sirtuin 2 ,medicine ,Escherichia coli ,Animals ,Humans ,Amino Acid Sequence ,HSP90 Heat-Shock Proteins ,RNA, Messenger ,RNA, Small Interfering ,Hormone response element ,Cell Nucleus ,biology ,Acetylation ,Cell Biology ,Original Articles ,Cell biology ,Rats ,030104 developmental biology ,chemistry ,Solubility ,030220 oncology & carcinogenesis ,biology.protein ,Molecular Medicine ,Original Article ,NAD+ kinase ,medicine.symptom ,Protein Binding ,Signal Transduction - Abstract
SIRT2 is a NAD+‐dependent deacetylase that deacetylates a diverse array of protein substrates and is involved in many cellular processes, including regulation of inflammation. However, its precise role in the inflammatory process has not completely been elucidated. Here, we identify heat‐shock protein 90α (Hsp90α) as novel substrate of SIRT2. Functional investigation suggests that Hsp90 is deacetylated by SIRT2, such that overexpression and knock‐down of SIRT2 altered the acetylation level of Hsp90. This subsequently resulted in disassociation of Hsp90 with glucocorticoid receptor (GR), and translocation of GR to the nucleus. This observation was further confirmed by glucocorticoid response element (GRE)‐driven reporter assay. Nuclear translocation of GR induced by SIRT2 overexpression repressed the expression of inflammatory cytokines, which were even more prominent under lipopolysaccharide (LPS) stimulation. Conversely, SIRT2 knock‐down resulted in the up‐regulation of cytokine expression. Mutation analysis indicated that deacetylation of Hsp90 at K294 is critical for SIRT2‐mediated regulation of cytokine expression. These data suggest that SIRT2 reduces the extent of LPS‐induced inflammation by suppressing the expression of inflammatory factors via SIRT2‐Hsp90‐GR axis.
- Published
- 2020
14. Baicalein Is a Phytohormone that Signals Through the Progesterone and Glucocorticoid Receptors
- Author
-
Rocío Rivera Rodríguez, Michael E. Johnson, Brenna J Kirkpatrick, Joanna E. Burdette, Julia R Austin, and Daniel D. Lantvit
- Subjects
Models, Molecular ,0301 basic medicine ,Agonist ,Cancer Research ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Mice, Nude ,Pharmacology ,Transfection ,Article ,Antioxidants ,Mice ,Random Allocation ,03 medical and health sciences ,chemistry.chemical_compound ,Receptors, Glucocorticoid ,0302 clinical medicine ,Endocrinology ,Wogonin ,Glucocorticoid receptor ,medicine ,Animals ,Humans ,Receptor ,Estrogen receptor activity ,Uncategorized ,Hormone response element ,biology ,Endocrine and Autonomic Systems ,Chemistry ,biology.organism_classification ,Baicalein ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Flavanones ,Scutellaria baicalensis ,Female ,Receptors, Progesterone ,Signal Transduction - Abstract
While flavonoids have been studied extensively for estrogen receptor activity, they have not been well studied for their ability to modify progesterone receptor (PR) and glucocorticoid receptor (GR) signaling. Three flavonoid compounds, tangeretin, wogonin, and baicalein, were selected for testing for PR and GR activity based on their structural similarity to known phytoprogesterone-like compounds. Each compound was docked in the binding pocket of PR and GR. Of these compounds, baicalein was predicted to be most likely to bind to both receptors. A fluorescence polarization competitive binding assay for PR and GR confirmed that baicalein binds to both the PR and GR with IC(50) values of 15.30 μM and 19.26 μM respectively. In Ishikawa PR-B and T47D cells, baicalein acted as a PR antagonist in a hormone response element (HRE) luciferase (Luc) assay. In OVCAR5 cells, which only express GR, baicalein was a GR agonist via an HRE/Luc assay and induced GR target genes, FKBP5 and GILZ. RU486, a PR and GR antagonist, abrogated baicalein’s activity in OVCAR5 cells, confirming baicalein’s activity is mediated through the GR. In vivo, baicalein administered intraperitoneally to female mice twice a week for 4 weeks at a dose of 25 mg/kg induced the GR target gene GILZ in the reproductive tract, which was blocked by RU486. In summary, baicalein has PR antagonist and GR agonist activity in vitro and demonstrates GR agonist activity in the uterus in vivo.
- Published
- 2020
15. Dominant Negative Activity by Estrogen and Progesterone Receptors
- Author
-
Yen, Paul M., Moudgil, V. K., editor, and Pavlik, Edward J., editor
- Published
- 1997
- Full Text
- View/download PDF
16. Progestins, Progesterone Receptors, and Breast Cancer
- Author
-
Horwitz, Kathryn B., Tung, Lin, Takimoto, Glenn S., Moudgil, V. K., editor, and Vedeckis, Wayne V., editor
- Published
- 1996
- Full Text
- View/download PDF
17. PPAR: a Key Nuclear Factor in Nutrient / Gene Interactions?
- Author
-
Desvergne, Béatrice, Wahli, Walter, Karin, Michael, editor, and Baeuerle, P. A., editor
- Published
- 1995
- Full Text
- View/download PDF
18. Estrogen Regulation of Uterine Proliferation: How Many ERRs Are Required?
- Author
-
Stancel, George M., Boettger-Tong, Holly L., Chiappetta, Connie, Hyder, Salman M., Kirkland, John L., Murthy, Lata, Loose-Mitchell, David S., and Dey, S. K., editor
- Published
- 1995
- Full Text
- View/download PDF
19. Mechanism of Action of Steroid Hormones and Antihormones: A Mini-overview
- Author
-
Baulieu, Etienne-Emile, Stock, Günter, editor, Habenicht, Ursula-F., editor, Chwalisz, K., editor, and Garfield, R. E., editor
- Published
- 1994
- Full Text
- View/download PDF
20. Hypothalamic Regulation of Corticotropin-Releasing Factor under Stress and Stress Resilience
- Author
-
Yasumasa Iwasaki, Makoto Daimon, and Kazunori Kageyama
- Subjects
Hypothalamo-Hypophyseal System ,medicine.medical_specialty ,endocrine system ,Corticotropin-Releasing Hormone ,QH301-705.5 ,Pituitary-Adrenal System ,Review ,Adrenocorticotropic hormone ,Biology ,Models, Biological ,Catalysis ,Inorganic Chemistry ,stress ,Adrenocorticotropic Hormone ,Anterior pituitary ,Stress, Physiological ,Internal medicine ,medicine ,Animals ,Humans ,Physical and Theoretical Chemistry ,hypothalamus ,Biology (General) ,Molecular Biology ,QD1-999 ,Spectroscopy ,Hormone response element ,Organic Chemistry ,corticotropin-releasing factor ,General Medicine ,Serum Response Element ,Adaptation, Physiological ,Computer Science Applications ,Chemistry ,Endocrinology ,Glucocorticoid secretion ,medicine.anatomical_structure ,Hypothalamus ,glucocorticoid ,FKBP5 ,Glucocorticoid ,hormones, hormone substitutes, and hormone antagonists ,medicine.drug - Abstract
This review addresses the molecular mechanisms of corticotropin-releasing factor (CRF) regulation in the hypothalamus under stress and stress resilience. CRF in the hypothalamus plays a central role in regulating the stress response. CRF stimulates adrenocorticotropic hormone (ACTH) release from the anterior pituitary. ACTH stimulates glucocorticoid secretion from the adrenal glands. Glucocorticoids are essential for stress coping, stress resilience, and homeostasis. The activated hypothalamic-pituitary-adrenal axis is suppressed by the negative feedback from glucocorticoids. Glucocorticoid-dependent repression of cAMP-stimulated Crf promoter activity is mediated by both the negative glucocorticoid response element and the serum response element. Conversely, the inducible cAMP-early repressor can suppress the stress response via inhibition of the cAMP-dependent Crf gene, as can the suppressor of cytokine signaling-3 in the hypothalamus. CRF receptor type 1 is mainly involved in a stress response, depression, anorexia, and seizure, while CRF receptor type 2 mediates “stress coping” mechanisms such as anxiolysis in the brain. Differential effects of FK506-binding immunophilins, FKBP4 and FKBP5, contribute to the efficiency of glucocorticoids under stress resilience. Together, a variety of factors contribute to stress resilience. All these factors would have the differential roles under stress resilience.
- Published
- 2021
21. The influence of estrogen receptor α signaling independent of the estrogen response element on avoidance behavior, social interactions, and palatable ingestive behavior in female mice
- Author
-
Troy A. Roepke, Jessica L. Verpeut, Daniel Regan, Patricia Ramirez, Kimberly Wiersielis, and Ali Yasrebi
- Subjects
Elevated plus maze ,medicine.medical_specialty ,medicine.drug_class ,Social Interaction ,Estrogen receptor ,Biology ,Response Elements ,Open field ,Article ,Behavioral Neuroscience ,Mice ,Endocrinology ,Gene knockin ,Internal medicine ,medicine ,Avoidance Learning ,Animals ,Hormone response element ,Mice, Knockout ,Behavior, Animal ,Estradiol ,Endocrine and Autonomic Systems ,Wild type ,Estrogen Receptor alpha ,Estrogens ,Feeding Behavior ,Social relation ,Estrogen ,Female - Abstract
Women are vulnerable to developing mental disorders that are associated with circulating estrogens. Estrogens, especially 17β-estradiol (E2), have a wide array of effects on the brain, affecting many behavioral endpoints associated with mental illness. By using a total estrogen receptor (ER) α knockout (KO), an ERα knock in/knock out (KIKO) that lacks a functional DNA-binding domain, and wild type (WT) controls treated with either oil or E2, we evaluated ERα signaling, dependent and independent of the estrogen response element (ERE), on avoidance behavior, social interactions and memory, and palatable ingestive behavior using the open field test, the elevated plus maze, the light dark box, the 3-chamber test, and palatable feeding. We found that ERα does not mediate control of anxiety-like behaviors but rather yielded differences in locomotor activity. In evaluating social preference and social recognition memory, we observed that E2 may modulate these measures in KIKO females but not KO females, suggesting that ERE-independent signaling is likely involved in sociability. Lastly, observations of palatable (high-fat) food intake suggested an increase in palatable eating behavior in oil-treated KIKO females. Oil-treated KO females had a longer latency to food intake, indicative of an anhedonic phenotype compared to oil-treated WT and KIKO females. We have observed that social-related behaviors are potentially influenced by ERE-independent ERα signaling and hedonic food intake requires signaling of ERα.
- Published
- 2021
22. Modulation of Hepatic and Renal Alcohol Dehydrogenase Activity and mRNA By Steroid Hormones in Vivo
- Author
-
Qulali, Mona, Dipple, Katrina M., Crabb, David W., Weiner, Henry, editor, Crabb, David W., editor, and Flynn, T. Geoffrey, editor
- Published
- 1993
- Full Text
- View/download PDF
23. Estrogen Receptor α Is Required for Maintaining Baseline Renin Expression.
- Author
-
Ko-Ting Lu, Keen, Henry L., Weatherford, Eric T., Sequeira-Lopez, Maria Luisa S., Gomez, R. Ariel, Sigmund, Curt D., and Lu, Ko-Ting
- Abstract
Enzymatic cleavage of angiotensinogen by renin represents the critical rate-limiting step in the production of angiotensin II, but the mechanisms regulating the initial expression of the renin gene remain incomplete. The purpose of this study is to unravel the molecular mechanism controlling renin expression. We identified a subset of nuclear receptors that exhibited an expression pattern similar to renin by reanalyzing a publicly available microarray data set. Expression of some of these nuclear receptors was similarly regulated as renin in response to physiological cues, which are known to regulate renin. Among these, only estrogen receptor α (ERα) and hepatic nuclear factor α have no known function in regulating renin expression. We determined that ERα is essential for the maintenance of renin expression by transfection of small interfering RNAs targeting Esr1, the gene encoding ERα, in renin-expressing As4.1 cells. We also observed that previously characterized negative regulators of renin expression, Nr2f2 and vitamin D receptor, exhibited elevated expression in response to ERα inhibition. Therefore, we tested whether ERα regulates renin expression through an interaction with Nr2f2 and vitamin D receptor. Renin expression did not return to baseline when we concurrently suppressed both Esr1 and Nr2f2 or Esr1 and vitamin D receptor mRNAs, strongly suggesting that Esr1 regulates renin expression independent of Nr2f2 and vitamin D receptor. ERα directly binds to the hormone response element within the renin enhancer region. We conclude that ERα is a previously unknown regulator of renin that directly binds to the renin enhancer hormone response element sequence and is critical in maintaining renin expression in renin-expressing As4.1 cells. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
24. Androgen receptor positively regulates gonadotropin-releasing hormone receptor in pituitary gonadotropes
- Author
-
Emily A. Witham, Shadi Shojaei, Emily V. Ho, Varykina G. Thackray, Stephanie C Bohaczuk, Genevieve E. Ryan, Jessica Cassin, and Pamela L. Mellon
- Subjects
0301 basic medicine ,Male ,Gonadotrophs ,Biochemistry ,Medical and Health Sciences ,LHRH ,Androgen ,Mice ,0302 clinical medicine ,Endocrinology ,Receptors ,Promoter Regions, Genetic ,GNRHR ,Biological Sciences ,Androgen receptor ,Receptors, Androgen ,Hormone receptor ,Androgens ,Chromatin Immunoprecipitation Sequencing ,Female ,Sequence Analysis ,Gonadotropin-releasing hormone receptor ,medicine.medical_specialty ,Heterologous ,030209 endocrinology & metabolism ,Biology ,Gonadotropic cell ,Article ,Cell Line ,Promoter Regions ,03 medical and health sciences ,Gonadotrope ,Endocrinology & Metabolism ,Genetic ,Internal medicine ,medicine ,Genetics ,Animals ,Molecular Biology ,Hormone response element ,Messenger RNA ,Agricultural and Veterinary Sciences ,Contraception/Reproduction ,GnRH receptor ,Neurosciences ,Sequence Analysis, DNA ,DNA ,030104 developmental biology ,Gene Expression Regulation ,Pituitary ,Receptors, LHRH - Abstract
Within pituitary gonadotropes, the gonadotropin-releasing hormone receptor (GnRHR) receives hypothalamic input from GnRH neurons that is critical for reproduction. Previous studies have suggested that androgens may regulate GnRHR, although the mechanisms remain unknown. In this study, we demonstrated that androgens positively regulate Gnrhr mRNA in mice. We then investigated the effects of androgens and androgen receptor (AR) on Gnrhr promoter activity in immortalized mouse LβT2 cells, which represent mature gonadotropes. We found that AR positively regulates the Gnrhr proximal promoter, and that this effect requires a hormone response element (HRE) half site at -159/-153 relative to the transcription start site. We also identified nonconsensus, full-length HREs at -499/-484 and -159/-144, which are both positively regulated by androgens on a heterologous promoter. Furthermore, AR associates with the Gnrhr promoter in ChIP. Altogether, we report that GnRHR is positively regulated by androgens through recruitment of AR to the Gnrhr proximal promoter.
- Published
- 2021
25. Estrogen Regulation of mRNAs in the Brain and Relationship to Lordosis Behavior
- Author
-
Lauber, A. H., Pfaff, D., Ganten, Detlev, editor, and Pfaff, Donald, editor
- Published
- 1990
- Full Text
- View/download PDF
26. LPS impairs steroidogenesis and ROS metabolism and induces PPAR transcriptional activity to disturb estrogen/androgen receptor expression in testicular cells
- Author
-
Gang Wang, Lingao Ju, Shanshan Zhang, Yu Xiao, Yuan Zhu, and Songtao Cheng
- Subjects
Lipopolysaccharides ,Male ,0301 basic medicine ,endocrine system ,Peroxisome Proliferator-Activated Receptors ,Gene Expression ,Peroxisome proliferator-activated receptor ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Spermatocytes ,Testis ,Genetics ,medicine ,Animals ,PPAR alpha ,Gonadal Steroid Hormones ,Spermatogenesis ,Molecular Biology ,Hormone response element ,chemistry.chemical_classification ,Sertoli Cells ,urogenital system ,Estrogens ,General Medicine ,Sertoli cell ,Spermatids ,Cell biology ,Mice, Inbred C57BL ,PPAR gamma ,Androgen receptor ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Receptors, Estrogen ,chemistry ,Receptors, Androgen ,Cell culture ,030220 oncology & carcinogenesis ,Androgens ,Reactive Oxygen Species ,Androgen Response Element - Abstract
Inflammation can deregulate the testicular functions of steroidogenesis and spermatogenesis, consequently contributing to male infertility. Animals and cells treated with lipopolysaccharide (LPS) exhibit infection- and inflammation-induced testicular dysfunction. However, the precise mechanisms affecting steroidogenesis and spermatogenesis in response to LPS-treatment remain poorly understood. We isolated distinct testicular cells including spermatocytes, round spermatids and late spermatids to analyze distribution of peroxisome proliferator-activated receptor (PPAR) family, plays central roles in the regulation of metabolism. Our results suggested Pparα/Pparγ mRNA was highly expressed in late spermatids, while Pparβ mRNA was highly expressed in round spermatids. To analyze the effect of LPS on testicular cells, we established an LPS infection model using primary Sertoli cells and testicular cell lines (TM4, GC2 and MLTC1). We observed that PPARγ and SIRT1 were concentrated in the nuclear region and that the mRNA expression levels of antioxidative enzymes (Cat and Homx1) and PPARγ were upregulated in primary Sertoli cells after LPS-treatment. Moreover, luciferase reporter gene assays of the testicular cell lines revealed that the activity of the PPAR response element (PPRE) was significantly increased. Importantly, the transcriptional activity of the androgen response element was significantly reduced, whereas activity of estrogen response element was strongly induced in LPS-treated TM4 cells, consistent with the RT-PCR results. Meanwhile, the qRT-PCR results revealed that the LPS-induced upregulation of Ar mRNA in MLTC1 cells and Erβ mRNA in TM4 cells were significantly recovered after treatment with the specific PPARγ-antagonist GW9662. In addition, we also found that LPS induced alterations in enzymes involved in steroidogenesis in testicular cell lines. Taken together, our results revealed that LPS may induce PPAR transcriptional activity to disturb estrogen/androgen receptor expression and impair steroidogenesis and ROS metabolism in testicular cells.
- Published
- 2019
27. Activation of hepatic estrogen receptor-α increases energy expenditure by stimulating the production of fibroblast growth factor 21 in female mice
- Author
-
Cristal M. Hill, Ellis R. Levin, Laurel A. Coons, Diana C. Albarado, Guy Fagherazzi, Kenneth S. Korach, John J. Lefante, Camille Allard, Franck Mauvais-Jarvis, Fabrice Bonnet, Christopher D. Morrison, and Beibei Xu
- Subjects
0301 basic medicine ,Aging ,FGF21 ,Physiology ,Estrogen receptor ,Endogeny ,Stimulation ,Cardiovascular ,Inbred C57BL ,Mice ,0302 clinical medicine ,ERα ,Mice, Knockout ,Metabolic syndrome ,Menopause ,Ovariectomized rat ,Original Article ,Female ,medicine.medical_specialty ,lcsh:Internal medicine ,medicine.drug_class ,Knockout ,030209 endocrinology & metabolism ,03 medical and health sciences ,Internal medicine ,Genetics ,medicine ,Animals ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,Metabolic and endocrine ,Nutrition ,Hormone response element ,business.industry ,Prevention ,Estrogen Receptor alpha ,Cell Biology ,medicine.disease ,Estrogen ,Fibroblast Growth Factors ,Mice, Inbred C57BL ,ER alpha ,030104 developmental biology ,Endocrinology ,Biochemistry and Cell Biology ,business ,Energy Metabolism - Abstract
Objective The endogenous estrogen 17β-estradiol (E2) promotes metabolic homeostasis in premenopausal women. In a mouse model of post-menopausal metabolic syndrome, we reported that estrogens increased energy expenditure, thus preventing estrogen deficiency-induced adiposity. Estrogens' prevention of fat accumulation was associated with increased serum concentrations of fibroblast growth factor 21 (FGF21), suggesting that FGF21 participates in estrogens' promotion of energy expenditure. Methods We studied the effect of E2 on FGF21 production and the role of FGF21 in E2 stimulation of energy expenditure and prevention of adiposity, using female estrogen receptor (ER)- and FGF21-deficient mice fed a normal chow and a cohort of ovariectomized women from the French E3N prospective cohort study. Results E2 acting on the hepatocyte ERα increases hepatic expression and production of FGF21 in female mice. In vivo activation of ERα increases the transcription of Fgf21 via an estrogen response element outside the promoter of Fgf21. Treatment with E2 increases oxygen consumption and energy expenditure and prevents whole body fat accumulation in ovariectomized female WT mice. The effect of E2 on energy expenditure is not observed in FGF21-deficient mice. While E2 treatment still prevents fat accumulation in FGF21-deficient mice, this effect is decreased compared to WT mice. In an observational cohort of ovariectomized women, E2 treatment was associated with lower serum FGF21 concentrations, which may reflect a healthier metabolic profile. Conclusions In female mice, E2 action on the hepatocyte ERα increases Fgf21 transcription and FGF21 production, thus promoting energy expenditure and partially decreasing fat accumulation., Highlights • Activation of the hepatocyte ERα increases FGF21 production in female mice. • ERα increases Fgf21 transcription in vivo via an estrogen response element. • Estrogen stimulation of energy expenditure is lost in female FGF21-deficient mice. • In women, estrogen treatment is associated with lower serum FGF21 concentrations.
- Published
- 2019
28. OTUD7B stabilizes estrogen receptor α and promotes breast cancer cell proliferation
- Author
-
Wei Chen, Gaosong Wu, Zelin Tian, Zeyu Wu, and Jianing Tang
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,Mice, Nude ,Estrogen receptor ,Breast Neoplasms ,Malignancy ,Article ,Cell growth ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Ovarian tumor ,Breast cancer ,0302 clinical medicine ,Cell Line, Tumor ,Endopeptidases ,Animals ,Humans ,Medicine ,skin and connective tissue diseases ,Gene ,Cell Proliferation ,Hormone response element ,Mice, Inbred BALB C ,QH573-671 ,Protein Stability ,business.industry ,Estrogen Receptor alpha ,Ubiquitination ,Cancer ,Cell Biology ,medicine.disease ,HEK293 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Cancer research ,Female ,Cytology ,business ,Protein Processing, Post-Translational ,Function (biology) - Abstract
Breast cancer is the most common malignancy in women worldwide. Estrogen receptor α (ERα) is expressed in ∼70% of breast cancer cases and promotes estrogen-dependent cancer progression. In the present study, we identified OTU domain-containing 7B (OTUD7B), a deubiquitylase belonging to A20 subgroup of ovarian tumor protein superfamily, as a bona fide deubiquitylase of ERα in breast cancer. OTUD7B expression was found to be positively correlated with ERα in breast cancer and associated with poor prognosis. OTUD7B could interact with, deubiquitylate, and stabilize ERα in a deubiquitylation activity-dependent manner. Depletion of OTUD7B decreased ERα protein level, the expression of ERα target genes, and the activity of estrogen response element in breast cancer cells. In addition, OTUD7B depletion significantly decreased ERα-positive breast cancer cell proliferation and migration. Finally, overexpression of ERα could rescue the suppressive effect induced by OTUD7B depletion, suggesting that the ERα status was essential to the function of OTUD7B in breast carcinogenesis. In conclusion, our study revealed an interesting post-translational mechanism between ERα and OTUD7B in ERα-positive breast cancer. Targeting the OTUD7B–ERα complex may prove to be a potential approach to treat patients with ERα-positive breast cancer.
- Published
- 2021
29. HNF4A is required to specify glucocorticoid action in the liver
- Author
-
David W. Ray, Andrew S. I. Loudon, Toryn Poolman, Frank J. Gonzalez, Mudassar Iqbal, Donghwan Kim, A. L. Hunter, and David A. Bechtold
- Subjects
Hormone response element ,Hepatocyte nuclear factors ,Glucocorticoid receptor ,Nuclear receptor ,Hepatocyte nuclear factor 4 ,Cistrome ,Chemistry ,medicine ,Context (language use) ,Glucocorticoid ,Cell biology ,medicine.drug - Abstract
The glucocorticoid receptor (GR) is a nuclear hormone receptor critical to the regulation of energy metabolism and the inflammatory response. The actions of GR are highly dependent on cell type and environmental context. Here, we demonstrate the necessity for liver lineage-determining factor hepatocyte nuclear factor 4A (HNF4A) in defining liver-specificity of GR action. In normal mouse liver, the HNF4 motif lies adjacent to the glucocorticoid response element (GRE) at GR binding sites found within regions of open chromatin. In the absence of HNF4A, the liver GR cistrome is remodelled, with both loss and gain of GR recruitment evident. Lost sites are characterised by HNF4 motifs and weak GRE motifs. Gained sites are characterised by strong GRE motifs, and typically show GR recruitment in non-liver tissues. The functional importance of these HNF4A-regulated GR sites is further demonstrated by evidence of an altered transcriptional response to glucocorticoid treatment in the Hnf4a-null liver.
- Published
- 2021
30. Nuclear receptor: Structure and function.
- Author
-
Sar P
- Subjects
- Humans, Ligands, Gene Expression, Cell Nucleus metabolism, Receptors, Cytoplasmic and Nuclear metabolism, Transcription Factors metabolism
- Abstract
Ligand-dependent transcription factors are nuclear receptors (NRs) that regulate various critical cellular processes such as reproduction, metabolism, development, etc. NRs are classified into (subgroup 0 to subgroup 6) seven superfamilies based on ligand-binding characteristics. All NRs share a general domain structure (A/B, C, D, and E) with distinct essential functions. NRs as monomers, homodimers, or heterodimers bind to consensus DNA sequences known as Hormone Response Elements (HREs). Furthermore, nuclear receptor-binding efficiency depends on minor differences in the sequences of HREs, spacing between the two half-sites, and the flanking sequence of the response elements. NRs can trans-activate and repress their target genes. In positively regulated genes, ligand-bound NRs recruit coactivators to activate the target gene expression, and unliganded NRs cause transcriptional repression. On the other hand, NRs repress gene expression by different mechanisms: (i) ligand-dependent transcriptional repression, (ii) ligand-independent transcriptional repression. This chapter will briefly explain NR superfamilies, their structures, molecular mechanism of action and their role in pathophysiological conditions, etc. That could enable the discovery of new receptors and their ligands and may elucidate their roles in various physiological processes. In addition, therapeutic agonists and antagonists would be developed to control the dysregulation of nuclear receptor signaling., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
31. Establishment of reporter cells that respond to glucocorticoids by a transposon-mediated promoter-trapping system
- Author
-
Kentaro Semba, Shinya Watanabe, Kosuke Ishikawa, and Sakura Tamamura
- Subjects
Hormone response element ,Transposable element ,Messenger RNA ,Clone (cell biology) ,Pharmaceutical Science ,02 engineering and technology ,Biology ,021001 nanoscience & nanotechnology ,Response Elements ,Transfection ,030226 pharmacology & pharmacy ,Dexamethasone ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,Glucocorticoid receptor ,Receptors, Glucocorticoid ,Rapid amplification of cDNA ends ,Genes, Reporter ,Luciferase ,0210 nano-technology ,Promoter Regions, Genetic ,Gene ,Glucocorticoids - Abstract
Previously, we had established a highly sensitive trap vector system for the efficient isolation of reporter cells for a certain condition of interest. In this study, we used this system to screen reporter cells that express the luciferase and enhanced green fluorescent protein genes in response to dexamethasone, a glucocorticoid receptor agonist to facilitate glucocorticoid signaling research. In total, 10 clones were isolated. The insertion sites of the trap vector were analyzed using 5' rapid amplification of cDNA ends (5' RACE), whereupon LPIN1, PKP2, and FKBP5 were identified as genes that were upregulated by the dexamethasone treatment. Specifically, PKP2 has not previously been focused as a gene that responds to glucocorticoids. The PKP2 mRNA was analyzed and induction of the endogenous gene was confirmed by real-time polymerase chain reaction. Given that PKP2 does not appear to have a consensus glucocorticoid response element (GRE) sequence, this reporter clone could supplement the current GRE-based reporter systems that are prevalently used. Because different clones showed different responses to glucocorticoids, these clones should provide more information than analysis with a single reporter clone. This paper demonstrates that the previously developed trap vector technology can contribute to the rapid construction of drug evaluation systems.
- Published
- 2020
32. Identification of estrogen receptor target genes involved in gonadal feminization caused by estrogen in Xenopus laevis
- Author
-
Xing-Hong Li, Jinbo Li, Yanping Shen, Yuan-Yuan Li, Zhanfen Qin, and Yiming Xiong
- Subjects
Hormone response element ,0303 health sciences ,biology ,medicine.drug_class ,Health, Toxicology and Mutagenesis ,Feminization (biology) ,Xenopus ,Estrogen receptor ,Promoter ,010501 environmental sciences ,Aquatic Science ,biology.organism_classification ,01 natural sciences ,Cell biology ,03 medical and health sciences ,Estrogen ,medicine ,Gene ,Chromatin immunoprecipitation ,hormones, hormone substitutes, and hormone antagonists ,030304 developmental biology ,0105 earth and related environmental sciences - Abstract
Estrogens and estrogenic endocrine disrupting chemicals can cause gonadal feminization in some vertebrates mainly through estrogen receptor (ER), but the underlying molecular mechanisms are unclear. The present study aimed to identify ER target genes involved in estrogen-caused gonadal feminization in Xenopus laevis. Based on our recent transcriptomic data that 10 nM 17β-estradiol (E2) altered gene transcription in feminizing gonads of male X. laevis at NF stages 48, 50, and 52, we searched estrogen response element (ERE) using the Dragon ERE Finder software in the promoter region of all the E2-regulated genes. As a result, 163 genes containing ERE sequence were identified as predicted ER target genes at NF stage 50 (on the 14th day postfertilization), a crucial stage for gonadal feminization. Then, some of these predicted ER target genes were further investigated, mainly including the genes that were suggested to be involved in E2-caused gonadal feminization and genes being dramatically up or down-regulated by E2. Fifteen genes were demonstrated to be responsive to E2, in turn ER antagonist blocked the E2-regulated transcription. Finally, we identified 10 genes that can bind to ERα by a chromatin immunoprecipitation-qPCR. Taken together, we identified the 10 genes that contain predicted ERE sequences, are responsive to estrogen and ER antagonist, and have ability to bind to ER as ER target genes, including pglyrp2, apoa1, fgb, tdo2, ca6, nags, cpb2, tmprss6, nudc, zwilch. Our results could help to improve the understanding of the molecular mechanisms for gonadal feminization caused by estrogenic endocrine disrupting chemicals in X. laevis, and even in other species.
- Published
- 2020
33. A steroid receptor coactivator acts as the DNA-binding partner of the methoprene-tolerant protein in regulating juvenile hormone response genes.
- Author
-
Li, Meng, Liu, Pengcheng, Wiley, Jessica D., Ojani, Reyhaneh, Bevan, David R., Li, Jianyong, and Zhu, Jinsong
- Subjects
- *
STEROID receptors , *DNA-binding proteins , *METHOPRENE , *JUVENILE hormones , *GENETIC regulation , *PROTEIN-protein interactions - Abstract
Methoprene-tolerant (Met) protein is a juvenile hormone (JH) receptor in insects. JH-bound Met forms a complex with the βFtz-F1-interacting steroid receptor coactivator (FISC) and together they regulate JH response genes in mosquitoes. Both proteins contain basic helix–loop–helix (bHLH) and PAS motifs. Here we demonstrated that FISC is the obligatory partner of Met for binding to JH-response elements (JHREs). Met or FISC alone could not bind a previously characterized JHRE, while formation of the Met–FISC complex was necessary and sufficient to bind to the JHRE. This binding required participation of the DNA-binding domains of both Met and FISC. The optimal DNA sequence recognized by Met and FISC contained a core consensus sequence GCACGTG. While formation of the Met–FISC complex in mosquito cells was induced by JH, heterodimerization and DNA binding of bacterially expressed Met and FISC were JH-independent, implying that additional mosquito proteins were required to modulate formation of the receptor complex. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
34. A satellite cell-specific knockout of the androgen receptor reveals myostatin as a direct androgen target in skeletal muscle.
- Author
-
Dubois, Vanessa, Laurent, Michaël R., Sinnesael, Mieke, Cielen, Nele, Helsen, Christine, Clinckemalie, Liesbeth, Spans, Lien, Gayan-Ramirez, Ghislaine, Deldicque, Louise, Hespel, Peter, Carmeliet, Geert, Vanderschueren, Dirk, and Claessens, Frank
- Subjects
- *
MUSCLES , *ANDROGENS , *SATELLITE cells , *CYTOLOGICAL research , *ANDROSTANE - Abstract
Androgens have well-established anabolic actions on skeletal muscle, although the direct effects of the androgen receptor (AR) in muscle remain unclear. We generated satellite cell-specific AR-knockout (satARKO) mice in which the AR is selectively ablated in satellite cells, the muscle precursor cells. Total-limb maximal grip strength is decreased by 7% in satARKO mice, with soleus muscles containing ~10% more type I fibers and 10% less type IIa fibers than the corresponding control littermates. The weight of the perineal levator ani muscle is markedly reduced (-52%). Thus, muscle AR is involved in fiber-type distribution and force production of the limb muscles, while it is a major determinant of the perineal muscle mass. Surprisingly,myostatin (Mstn), a strong inhibitor of skeletal muscle growth, is one of the most androgen-responsive genes (6-fold reduction in satARKO) through direct transcription activation by the AR. Consequently, muscle hypertrophy in response to androgens is augmented in Mstn-knockout mice. Our finding that androgens induce Mstn signaling to restrain their own anabolic actions has implications for the treatment of muscle wasting disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
35. Phlpp1 is induced by estrogen in osteoclasts and its loss in Ctsk-expressing cells does not protect against ovariectomy-induced bone loss
- Author
-
Marcelline K. Hanson, David H. H. Molstad, Elizabeth W. Bradley, Kim C. Mansky, Andrew Norton, and Ismael Y. Karkache
- Subjects
0301 basic medicine ,Physiology ,Cathepsin K ,Osteoclasts ,Gene Expression ,Biochemistry ,Mice ,0302 clinical medicine ,Animal Cells ,Gene expression ,Phosphoprotein Phosphatases ,Medicine and Health Sciences ,Lipid Hormones ,Reproductive System Procedures ,Musculoskeletal System ,Connective Tissue Cells ,Mice, Knockout ,Multidisciplinary ,Estradiol ,Chemistry ,Genetically Modified Organisms ,Cell Differentiation ,medicine.anatomical_structure ,Connective Tissue ,Medicine ,Immunohistochemistry ,Engineering and Technology ,Female ,Bone Remodeling ,Anatomy ,Cellular Types ,Genetic Engineering ,Research Article ,Biotechnology ,medicine.medical_specialty ,medicine.drug_class ,Science ,Ovariectomy ,Phosphatase ,Surgical and Invasive Medical Procedures ,Bioengineering ,Bone resorption ,03 medical and health sciences ,Osteoclast ,In vivo ,Internal medicine ,medicine ,Genetics ,Animals ,Bone Resorption ,Bone ,Skeleton ,Hormone response element ,Osteoblasts ,Surgical Excision ,Genetically Modified Animals ,Biology and Life Sciences ,Estrogens ,Cell Biology ,Hormones ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Biological Tissue ,Insulin-Like Growth Factor Binding Protein 4 ,Estrogen ,Osteoporosis ,Physiological Processes ,030217 neurology & neurosurgery - Abstract
Prior studies demonstrated that deletion of the protein phosphatase Phlpp1 in Ctsk-Cre expressing cells enhances bone mass, characterized by diminished osteoclast activity and increased coupling to bone formation. Due to non-specific expression of Ctsk-Cre, the definitive mechanism for this observation was unclear. To further define the role of bone resorbing osteoclasts, we performed ovariectomy (Ovx) and Sham surgeries on Phlpp1 cKOCtsk and WT mice. Micro-CT analyses confirmed enhanced bone mass of Phlpp1 cKOCtsk Sham females. In contrast, Ovx induced bone loss in both groups, with no difference between Phlpp1 cKOCtsk and WT mice. Histomorphometry demonstrated that Ovx mice lacked differences in osteoclasts per bone surface, suggesting that estradiol (E2) is required for Phlpp1 deficiency to have an effect. We performed high throughput unbiased transcriptional profiling of Phlpp1 cKOCtsk osteoclasts and identified 290 differentially expressed genes. By cross-referencing these differentially expressed genes with all estrogen response element (ERE) containing genes, we identified IGFBP4 as potential estrogen-dependent target of Phlpp1. E2 induced PHLPP1 expression, but reduced IGFBP4 levels. Moreover, genetic deletion or chemical inhibition of Phlpp1 was correlated with IGFBP4 levels. We then assessed IGFBP4 expression by osteoclasts in vivo within intact 12-week-old females. Modest IGFBP4 immunohistochemical staining of TRAP+ osteoclasts within WT females was observed. In contrast, TRAP+ bone lining cells within intact Phlpp1 cKOCtsk females robustly expressed IGFBP4, but levels were diminished within TRAP+ bone lining cells following Ovx. These results demonstrate that effects of Phlpp1 conditional deficiency are lost following Ovx, potentially due to estrogen-dependent regulation of IGFBP4.
- Published
- 2020
36. Repression of transcription by the glucocorticoid receptor: A parsimonious model for the genomics era
- Author
-
Anthony N. Gerber, Robert Newton, and Sarah K. Sasse
- Subjects
0301 basic medicine ,Transcription, Genetic ,nGRE, negative glucocorticoid response element ,Anti-Inflammatory Agents ,Biology ,Biochemistry ,Dexamethasone ,03 medical and health sciences ,Glucocorticoid receptor ,Receptors, Glucocorticoid ,TSS, transcriptional start site ,Transcription (biology) ,GRE, glucocorticoid response element ,glucocorticoid receptor ,Animals ,Humans ,Enhancer ,Molecular Biology ,Transcription factor ,Psychological repression ,Glucocorticoids ,Hormone response element ,Regulation of gene expression ,Inflammation ,GR, glucocorticoid receptor ,030102 biochemistry & molecular biology ,Models, Genetic ,SARS-CoV-2 ,JBC Reviews ,NF-kappa B ,COVID-19 ,Cell Biology ,Genomics ,negative feedback ,Cell biology ,COVID-19 Drug Treatment ,transcriptional enhancer ,030104 developmental biology ,Gene Expression Regulation ,Signal transduction ,repression ,Signal Transduction - Abstract
Glucocorticoids are potent anti-inflammatory drugs that are used to treat an extraordinary range of human disease, including COVID-19, underscoring the ongoing importance of understanding their molecular mechanisms. Early studies of GR signaling led to broad acceptance of models in which glucocorticoid receptor (GR) monomers tether repressively to inflammatory transcription factors, thus abrogating inflammatory gene expression. However, newer data challenge this core concept and present an exciting opportunity to reframe our understanding of GR signaling. Here, we present an alternate, two-part model for transcriptional repression by glucocorticoids. First, widespread GR-mediated induction of transcription results in rapid, primary repression of inflammatory gene transcription and associated enhancers through competition-based mechanisms. Second, a subset of GR-induced genes, including targets that are regulated in coordination with inflammatory transcription factors such as NF-κB, exerts secondary repressive effects on inflammatory gene expression. Within this framework, emerging data indicate that the gene set regulated through the cooperative convergence of GR and NF-κB signaling is central to the broad clinical effectiveness of glucocorticoids in terminating inflammation and promoting tissue repair.
- Published
- 2020
37. TRIIODOTHYRONINE ACTIVATES GLYCEROL-3-PHOSPHATE ACYLTRANSFERASE 3 VIA AGGTCA-LIKE-DIRECT-REPEAT-4 TYPE THYROID HORMONE RESPONSE ELEMENT
- Author
-
Baoling Han, Y Zhang, J Han, G Y Liu, Jian Wang, Lifeng Zhao, Xia Jiang, and Y Iwasaki
- Subjects
Hormone response element ,Triiodothyronine ,Endocrine and Autonomic Systems ,business.industry ,Endocrinology, Diabetes and Metabolism ,Thyroid ,Promoter ,Retinoid X receptor ,Molecular biology ,Endocrinology ,medicine.anatomical_structure ,Acyltransferase ,Medicine ,Luciferase ,Electrophoretic mobility shift assay ,business ,General Endocrinology - Abstract
Background Thyroid hormone participates in lipid metabolism regulation. However, the effects on triacyleride or triacylglycerol metabolism are complex and not fully clarified yet. In this study, we try to identify novel thyroid hormone-targeting lipogenic metabolic genes and analyze their molecular regulative mechanism. Method Thirty-five promoters of twenty-nine human lipogenic regulative enzyme genes were constructed into pXP1 luciferase reporter plasmid (PFK2/FBP2-luc) and transfected into HeGP2 cells, respectively. Gene expression induced by triiodothyronine (T3) was detected by luciferase assay. The T3-activated gene promoter was then analyzed by sequence analysis, deletion and mutation, and electrophoretic mobility shift assay (EMSA). Results After 10 nM T3 stimulation for 36 h, phosphogluconate dehydrogenase, malic enzyme, Glycerol-3-phosphate acyltransferase (GPAT) 3, and 1-acylglycerol-3-phosphate O-acyltransferase (AGPAT) 2 were significantly activated, respectively. A AGGTCA-like-direct-repeat-4 consensus thyroid hormone response element (DR4-TRE)-like sequence was found in the GPAT3 promoter, which was then verified to be necessary for T3-induced GPAT3 activation by gene deletion and mutation analysis. EMSA further identified that T3-thyroid receptor (TR) α-retinoid-X receptor (RXR) complex directly bound on the GPAT3 promoter. Conclusion Triiodothyronine could activate the GPAT3 through DR4-TRE-like sequence binding to participate in lipogenic regulation. AGPAT2 may be another thyroid hormone target enzyme.
- Published
- 2020
38. Estrogen-ERα signaling and DNA hypomethylation co-regulate expression of stem cell protein PIWIL1 in ERα-positive endometrial cancer cells
- Author
-
Hua-Jing Yang, Xiaoping Wan, Minjiao Zhu, Qin Lin, Zheng Chen, Yingying Yu, and Xiaoying He
- Subjects
Carcinogenesis ,lcsh:Medicine ,Endometrial carcinoma ,Biology ,medicine.disease_cause ,Biochemistry ,Small hairpin RNA ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,lcsh:QH573-671 ,Molecular Biology ,Cell Proliferation ,ERα ,030304 developmental biology ,Hormone response element ,0303 health sciences ,DNA methylation ,Oncogene ,lcsh:Cytology ,Research ,Endometrial cancer ,lcsh:R ,Estrogen Receptor alpha ,Estrogens ,Cell Biology ,medicine.disease ,Endometrial Neoplasms ,Gene Expression Regulation, Neoplastic ,PIWIL1 ,030220 oncology & carcinogenesis ,Argonaute Proteins ,Cancer cell ,Cancer research ,Female ,Chromatin immunoprecipitation - Abstract
Background We previously identified PIWIL1 as an oncogene involved in endometrial carcinogenesis. However, the mechanism of Piwil1 mediated regulation of tumorigenesis remains poorly understood. Methods The expression levels of target genes in endometrial cancer cells were detected by quantitative reverse transcription-PCR (RT-qPCR) and western blotting. Up- or down-regulation of ERα or PIWIL1 was achieved by transient transfection with expressing plasmids or short hairpin RNA (shRNA). Dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) were used to demonstrate the ERα bound to the half estrogen response element (half-ERE) located in PIWIL1 promoter. The expression of PIWIL1 and ERα in endometrial carcinoma tissues were investigated using immunohistochemistry and RT-qPCR. The proliferation ability of cancer cells were evaluated by MTT. Methylation status of the PIWIL1 promoter was detected by bisulfite sequencing PCR (BSP). Results In the present study, we found that PIWIL1 mediated E2-stimulated cancer cell proliferation. In ERα-positive endometrial cancer cells, we demonstrated that estrogen-ERα signaling significantly up-regulated the expression of PIWIL1, which was mediated by binding of the ERα onto the PIWIL1 promoter. Furthermore, we found that a half-ERE in the PIWIL1 promoter was essential for ERα binding. The PIWIL1 promoter was hypomethylated in ERα-positive endometrial cancer cells. Treatment with 5-aza-deoxycytidine (5-aza-dC) could up-regulate PIWIL1 expression. Conclusions These findings uncover a novel molecular mechanism by which estrogen-ERα signaling and DNA hypomethylation co-regulate PIWIL1 expression. These findings provide novel insights into the hormonal regulation of PIWIL1 in endometrial cancer and the PIWIL1’s role in estrogen-stimulated endometrial carcinogenesis.
- Published
- 2020
39. Determining the Fasting Length in Time-Restricted Feeding: Analysis of Endocrine Function Among Young, Healthy, Normal Weight, European Men
- Author
-
Wasantha Jayawardene, Stephanie L. Dickinson, Xiwei Chen, Ann E. K. Kosobud, and Stephen J. Carter
- Subjects
Hormone response element ,medicine.medical_specialty ,Nutrition and Dietetics ,Adiponectin ,medicine.diagnostic_test ,business.industry ,C-peptide ,Insulin ,medicine.medical_treatment ,Medicine (miscellaneous) ,chemistry.chemical_compound ,Endocrinology ,chemistry ,Gluconeogenesis ,Internal medicine ,Immunoassay ,Medicine ,Endocrine system ,business ,Eating Frequency and Chrononutrition ,Food Science ,Hydrocortisone ,medicine.drug - Abstract
OBJECTIVES: Intermittent calorie restriction has emerged as an effective alternative to continuous calorie restriction to impart favorable health benefits. However, there remains a degree of uncertainty about the optimal fasting length to simultaneously improve metabolic flexibility and promote positive behavior-change. While a 16-hr fast is commonly employed, it is possible that a shorter fast could result in similar metabolic benefits and better longer term adherence. Thus, we compared changes in hormone response between two 4-hr time intervals of fasting – 11–15 hrs. vs. 15–19 hrs. METHODS: Secondary analyses were performed from a 38-hr fasting study that enrolled 24 healthy, European men (18–30 y) with normal body mass index. Participants initiated a fast at 2300, arrived to the laboratory at 0800, and continued fasting in individual rooms. Venous blood was sampled at 13 time-points to measure insulin, glucose, adrenocorticotropin (ACTH), C-peptide, cortisol, and adiponectin via immunoassays. Data were normally distributed without outliers or missing values. Within-group differences were analyzed with paired t-tests. RESULTS: Insulin (pmol/L) declined between 11–15 hrs. (7.00; CI = 0.97, 13.02; P = .025), but not 15–19 hrs. (5.67; CI = –0.65, 11.98; P = .076). The decline in C-peptide (nmol/L) was sharper between 11–15 hrs. (0.09; CI = 0.05, 0.13; P = < .001), compared to 15–19 hrs. (0.06; CI = 0.02, 0.09; P = .003). Cortisol (nmol/L) also steeply declined between 11–15 hrs. (60.09; CI = 30.58, 89.59; P = < .001), but slightly increased between 15–19 hrs. (10.75; CI = −33.15, 11.65; P = 0.331). The change in glucose (mmol/L) was not different between 11–15 hrs. (0.02 mmol/L; CI = −0.15, 0.19; P = 0.784), but sharply decreased between 15–19 hrs. (0.24; CI = 0.10, 0.39; P = 0.002). No differences were detected for ACTH or adiponectin. CONCLUSIONS: Though substrate oxidation was not measured, changes in insulin, C-peptide, cortisol, and glucose were consistent with increased gluconeogenesis during 11–15 hrs. and lipid oxidation during 15–19 hrs. of fasting. Whereas cortisol often declines throughout the day, a slight increase was noted between 15–19 hrs. – evidence of a heightened sympathetic activity. Further study of hormone response during intermittent fasting will inform optimal length to promote health benefits and longer term adherence. FUNDING SOURCES: None
- Published
- 2020
40. Protein Network Studies on PCOS Biomarkers With S100A8, Druggability Assessment, and RNA Aptamer Designing to Control Its Cyst Migration Effect
- Author
-
Madasamy Swathi, Renganathan Venkatalakshmi, Manickam Kiruthika, Ayyachamy Shobana, K Suhasini, Anant Achary, Sharon Roopathy, Kandasamy Thirukumaran, and Subramaniyan Manibalan
- Subjects
0301 basic medicine ,Histology ,In silico ,pcos targets ,lcsh:Biotechnology ,Druggability ,Biomedical Engineering ,Bioengineering ,02 engineering and technology ,Biology ,lim method ,S100A8 ,03 medical and health sciences ,lcsh:TP248.13-248.65 ,network analysis ,Original Research ,Hormone response element ,protein network ,RNA ,Bioengineering and Biotechnology ,021001 nanoscience & nanotechnology ,Polycystic ovary ,030104 developmental biology ,RNA aptamer ,Lipofectamine ,Cancer research ,Target protein ,0210 nano-technology ,druggability ,Biotechnology - Abstract
The prevalence of polycystic ovary syndrome (PCOS) has been gradually increasing among adult females worldwide. Laparoscopy drilling on ovary is the only available temporary solution with a high incidence of reoccurrence. S100A8 with S100A9 complex is believed to facilitate the cyst migration in PCOS condition. The high evident protein interaction network studies between PCOS biomarkers, cancer invasion markers, and the interactors of S100A8 confirm that this protein has strong interaction with other selective PCOS biomarkers, which may be associative in the immature cyst invasion process. Through the network studies, intensive structural and pathway analysis, S100A8 is identified as a targetable protein. In this research, the non-SELEX in silico method is adapted to construct RNA Library based on the consensus DNA sequence of Glucocorticoid Response Element (GRE) and screened the best nucleotide fragments which are bound within the active sites of the target protein. Selected sequences are joined as a single strand and screened the one which competitively binds with minimal energy. In vitro follow-up of this computational research, the designed RNA aptamer was used to infect the MCF7 cell line through Lipofectamine 2000 mediated delivery to study the anti-cell migration effect. Wound Scratch assay confirms that the synthesized 18-mer oligo has significant inhibition activity toward tumor cell migration at the cellular level.
- Published
- 2020
41. TRIM11 promotes breast cancer cell proliferation by stabilizing estrogen receptor α
- Author
-
Jianing Tang, Gaosong Wu, Yongwen Luo, Qian Yang, Qiuxia Cui, Zelin Tian, and Xing Liao
- Subjects
0301 basic medicine ,Cancer Research ,Original article ,Ubiquitin-Protein Ligases ,Estrogen receptor ,Apoptosis ,Breast Neoplasms ,Biology ,lcsh:RC254-282 ,Tripartite Motif Proteins ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Polyubiquitination ,Ubiquitin ,Cell Movement ,medicine ,Biomarkers, Tumor ,Tumor Cells, Cultured ,Humans ,Cell Proliferation ,ERα ,TRIM11 ,Hormone response element ,Cell growth ,Estrogen Receptor alpha ,Cancer ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cytoplasm ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Female ,Mono-ubiquitination - Abstract
Breast cancer is the most commonly diagnosed malignancy in female worldwide, over 70% of which are estrogen receptor α (ERα) positive. ERα has a crucial role in the initiation and progression of breast cancer and is an indicator of endocrine therapy, while endocrine resistance is an urgent problem in ER-positive breast cancer patients. In the present study, we identify a novel E3 ubiquitin ligase TRIM11 function to facilitate ERα signaling. TRIM11 is overexpressed in human breast cancer, and associates with poor prognosis. The protein level of TRIM11 is highly correlated with ERα. RNA-seq results suggest that ERα signaling may be an underlying target of TRIM11. Depletion of TRIM11 in breast cancer cells significantly decreases cell proliferation and migration. And the suppression effects can be reversed by overexpressing ERα. In addition, ERα protein level, ERα target genes expression and estrogen response element activity are also dramatically decreased by TRIM11 depletion. Further mechanistic analysis indicates that the RING domain of TRIM11 interacted with the N terminal of ERα in the cytoplasm and promotes its mono-ubiquitination, thus enhances ERα protein stability. Our study describes TRIM11 as a modulating factor of ERα and increases ERα stability via mono-ubiquitination. TRIM11 could be a promising therapeutic target for breast cancer treatment.
- Published
- 2020
42. Regulatory Sharing Between Estrogen Receptor α Bound Enhancers
- Author
-
Julia B. Carleton, Jason Gertz, Ryan M. Layer, Aaron R. Quinlan, Matthew Ginley-Hidinger, and Kristofer C. Berrett
- Subjects
Transcription, Genetic ,AcademicSubjects/SCI00010 ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Genetics ,Humans ,Nucleotide Motifs ,Promoter Regions, Genetic ,Enhancer ,Gene ,030304 developmental biology ,Hormone response element ,Regulation of gene expression ,0303 health sciences ,biology ,Genome, Human ,Chemistry ,Gene regulation, Chromatin and Epigenetics ,Estrogen Receptor alpha ,Acetylation ,Promoter ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Enhancer Elements, Genetic ,Histone ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,biology.protein ,030217 neurology & neurosurgery ,hormones, hormone substitutes, and hormone antagonists - Abstract
The human genome encodes an order of magnitude more gene expression enhancers than promoters, suggesting that most genes are regulated by the combined action of multiple enhancers. We have previously shown that neighboring estrogen-responsive enhancers, which are approximately 5,000 basepairs apart, exhibit complex synergistic contributions to the production of an estrogenic transcriptional response. Here we sought to determine the molecular underpinnings of the observed enhancer cooperativity. We generated genetic deletions of individual estrogen receptor α (ER) bound enhancers and found that enhancers containing full estrogen response element (ERE) motifs control ER binding at neighboring sites, while enhancers with pre-existing histone acetylation/accessibility confer a permissible chromatin environment to the neighboring enhancers. Genome engineering revealed that a cluster of two enhancers with half EREs could not compensate for the lack of a full ERE site within the cluster. In contrast, two enhancers with full EREs produced a transcriptional response greater than the wild-type locus. By swapping genomic sequences between enhancers, we found that the genomic location in which a full ERE resides strongly influences enhancer activity. Our results lead to a model in which a full ERE is required for ER recruitment, but the presence of a pre-existing active chromatin environment within an enhancer cluster is also needed in order for estrogen-driven gene regulation to occur.
- Published
- 2020
43. Endothelial cell–glucocorticoid receptor interactions and regulation of Wnt signaling
- Author
-
Sameet Mehta, Carlos Fernández-Hernando, William C. Sessa, Xinbo Zhang, Han Zhou, Swayam Prakash Srivastava, Ahmad F. Hedayat, Kariona A. Grabińska, Paola Perrotta, Chris Wong, and Julie E. Goodwin
- Subjects
0301 basic medicine ,Male ,Vasculitis ,Chromatin Immunoprecipitation ,Endogeny ,Inflammation ,Biology ,Dexamethasone ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Glucocorticoid receptor ,Receptors, Glucocorticoid ,Downregulation and upregulation ,medicine ,Animals ,Wnt Signaling Pathway ,Hormone response element ,Mice, Knockout ,Wnt signaling pathway ,Intracellular Signaling Peptides and Proteins ,General Medicine ,DNA ,Cell biology ,Endothelial stem cell ,030104 developmental biology ,Frzb ,030220 oncology & carcinogenesis ,Multigene Family ,Endothelium, Vascular ,medicine.symptom ,hormones, hormone substitutes, and hormone antagonists ,Research Article ,Protein Binding - Abstract
Vascular inflammation is present in many cardiovascular diseases, and exogenous glucocorticoids have traditionally been used as a therapy to suppress inflammation. However, recent data have shown that endogenous glucocorticoids, acting through the endothelial glucocorticoid receptor, act as negative regulators of inflammation. Here, we performed ChIP for the glucocorticoid receptor, followed by next-generation sequencing in mouse endothelial cells to investigate how the endothelial glucocorticoid receptor regulates vascular inflammation. We identified a role of the Wnt signaling pathway in this setting and show that loss of the endothelial glucocorticoid receptor results in upregulation of Wnt signaling both in vitro and in vivo using our validated mouse model. Furthermore, we demonstrate glucocorticoid receptor regulation of a key gene in the Wnt pathway, Frzb, via a glucocorticoid response element gleaned from our genomic data. These results suggest a role for endothelial Wnt signaling modulation in states of vascular inflammation.
- Published
- 2020
44. A NOVEL GLUCOCORTICOID RECEPTOR MUTATION IN PRIMARY GENERALIZED GLUCOCORTICOID RESISTANCE DISEASE
- Author
-
Yang Long, Ji De, Zhen Xiao, Xiaozhen Tan, Tao Chen, Haoming Tian, Jiaqi Li, Yan Ren, and Lifen Ma
- Subjects
Male ,medicine.medical_specialty ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Mutant ,030209 endocrinology & metabolism ,Adrenocorticotropic hormone ,Dexamethasone ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Endocrinology ,Glucocorticoid receptor ,Receptors, Glucocorticoid ,Internal medicine ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,030212 general & internal medicine ,Receptor ,Glucocorticoids ,Hormone response element ,business.industry ,Mutation ,business ,Glucocorticoid ,Metabolism, Inborn Errors ,Genetic screen ,medicine.drug - Abstract
Objective: Primary generalized glucocorticoid resistance (PGGR) is a rare hereditary disease characterized by generalized partial target-tissue insensitivity to glucocorticoids. To date, few cases have been reported, and more cases, especially involving other races, are needed to fully understand this disease. Methods: This study presented a novel glucocorticoid receptor mutation in a PGGR pedigree. The index patient was a 14-year-old male with fatigue, hypokalemia, hypertension, and polyuria. Eleven family members were available for the genetic screen. Next-generation sequencing and Sanger sequencing were used to identify the mutation. We systematically investigated the molecular mechanism through which the mutation impaired glucocorticoid signal transduction in COS-7 cells. Results: The index patient carried a de novo homo-zygous mutation within exon 6 (c.1652C>A, p.551S>Y), whereas eight family members carrying a heterozygous mutation were all phenotypically silent. The affinity of the human glucocorticoid receptor (hGR) for the ligand was 1.97-fold lower in the patient than in the family members. Mutant hGRα (551Y) displayed a 3.2-fold reduction in its ability to transactivate glucocorticoid-responsive genes. When exposed to the same concentration of dexamethasone, hGRα (551Y) displayed a reduced ability to trans-locate into the nucleus and decreased levels of hGR dimer formation and could not effectively induce the glucocorticoid response element to regulate the transcription of related genes. After 2 years of dexamethasone treatment, the volume of the left and right adrenal glands of the index subject decreased by 55.6% and 32.4%, respectively. The pituitary volume decreased by 18.9%. During the 2-year follow-up, none of the heterozygous carriers developed hypertension or hypokalemia. Conclusion: We described a novel homozygous glucocorticoid receptor mutation causing PGGR. This homozygous mutation leads to hypertension and hypokalemia, but its heterozygous mutation has no relevant clinical symptoms. Abbreviations: ACTH = adrenocorticotropic hormone; DBD = DNA-binding domain; GR = glucocorticoid receptor; GRE = glucocorticoid response element; hGR = human glucocorticoid receptor; LBD = ligand-binding domain; PGGR = primary generalized glucocorticoid resistance.
- Published
- 2020
45. Human Peroxisomal 3-Ketoacyl-CoA Thiolase: Tissue Expression and Metabolic Regulation
- Author
-
Norbert Latruffe
- Subjects
Hormone response element ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Chemistry ,Thiolase ,Response element ,030212 general & internal medicine ,Binding site ,Peroxisome ,Molecular biology ,Gene ,Transcription factor - Abstract
This paper reports that the human peroxisomal 3-ketoacyl-CoA thiolase expression shows three transcripts: Tr1 (1705 bp), Tr2 (1375 bp) and Tr3 (1782 bp). Their highest expression is observed in the human liver and at a lesser extent in hepatic-derived HepG2 cells. The intestine and blood and endothelial cells show lower expression. The lowest expression is found in adipocytes. The transcript Tr3 appears to be the most abundant. So far, no data have been published regarding the regulation of the human peroxisomal thiolase. After cloning a fragment of the 5′ region involved in the regulation of the human thiolase gene, the effects of different treatments have been studied on the thiolase expression in the hepatoma HepG2 human cell line. Biocomputing analysis indicates that (i) a GRE (glucocorticoid response element) is located at −650 bp upstream of the transcription initiation site; (ii) a C/EBPα (CCAAT/enhancer-binding protein) binding site is located at − 1000 bp upstream of the transcription initiation site – and (iii) there is no putative PPRE (peroxisome proliferator-activated receptor response element). In the human HepG2 cells, thiolase expression is upregulated by glucose and downregulated by insulin and sterols, while dexamethasone and fatty acids have no effect. The ciprofibrate, a peroxisome proliferator, leads only to a weak stimulation of the mRNA expression as compared to thiolase B expression in the rat liver.
- Published
- 2020
46. 17β-estradiol binding to ERα promotes the progression of prolactinoma through estrogen-response element-induced CaBP-9k up-regulation
- Author
-
Hao Han, Jun Liu, Gaoyang Fan, and Wenpeng Lu
- Subjects
Cell Survival ,medicine.drug_class ,Biophysics ,Down-Regulation ,Estrogen receptor ,Apoptosis ,Response Elements ,Biochemistry ,Prolactin cell ,Small hairpin RNA ,S100 Calcium Binding Protein G ,Cell Line, Tumor ,medicine ,Animals ,Immunoprecipitation ,RNA, Messenger ,Viability assay ,Molecular Biology ,Research Articles ,Cell Proliferation ,Cancer ,ERα ,Hormone response element ,Estradiol ,Chemistry ,Estrogen Receptor alpha ,Estrogens ,Cell Biology ,Transfection ,CaBP-9k ,ERE ,Estradiol binding ,Rats ,Up-Regulation ,Cell biology ,Estrogen ,prolactinoma ,Cell Cycle, Growth & Proliferation ,17β-estradiol (E2) ,hormones, hormone substitutes, and hormone antagonists - Abstract
17β-estradiol (E2) is considered to be an important instigator of prolactinoma, and can positively regulate the expression of calbindin-D9k (CaBP-9k) which contains an estrogen responsive element (ERE) via estrogen receptors (ERs). However, the detailed mechanism of E2 in promoting CaBP-9k expression and their roles in prolactinoma progression remain unclear. Here, we aimed to characterize it. The luciferase gene reporter assay with luc-ERE transfection showed that E2 treatment significantly enhanced the transcriptional level of CaBP-9k, whereas CaBP-9k activity was reduced when GH3 and MMQ cells were treated with AZD9496, an antagonist of ERα. E2 treatment increased the protein expressions of CaBP-9k and ERα but not ERβ, whereas this effect was also abolished when cells were treated with AZD9496. Besides, immunoprecipitation (IP) and immunofluorescence assays demonstrated that CaBP-9k could directly interact with ERα not ERβ, and Chromatin IP (ChIP) assay showed that ERα could bind to ERE of the CaBP-9k promoter. Moreover, cell counting kit-8 (CCK-8) and flow cytometry assays showed that E2 treatment significantly enhanced cell viability and inhibited cell apoptosis, but these effects were all abolished when ERα was down-regulated by short hairpin RNA (shRNA) or inhibited by AZD9496, as well as CaBP-9K suppression in both GH3 and MMQ cell lines. Taken together, these findings indicated that E2 stimulation promoted prolactin cell proliferation and inhibited cell apoptosis through ERα-induced CaBP-9k up-regulation, which then accelerated the advanced progression of prolactinoma.
- Published
- 2020
47. Polybutylcyanoacrylate nanoparticles for delivering hormone response element-conjugated neurotrophin-3 to the brain of intracerebral hemorrhagic rats.
- Author
-
Chung, Chiu-Yen, Yang, Jen-Tsung, and Kuo, Yung-Chih
- Subjects
- *
CEREBRAL hemorrhage , *ACRYLATES , *NANOMEDICINE , *HORMONE therapy , *LABORATORY rats , *DRUG delivery systems , *NEUROTROPHINS - Abstract
Abstract: Hypertensive intracerebral hemorrhage (ICH) is a rapidly evolutional pathology, inducing necrotic cell death followed by apoptosis, and alters gene expression levels in surrounding tissue of an injured brain. For ICH therapy by controlled gene release, the development of intravenously administrable delivery vectors to promote the penetration across the blood–brain barrier (BBB) is a critical challenge. To enhance transfer efficiency of genetic materials under hypoxic conditions, polybutylcyanoacrylate (PBCA) nanoparticles (NPs) were used to mediate the intracellular transport of plasmid neurotrophin-3 (NT-3) containing hormone response element (HRE) with a cytomegalovirus (cmv) promoter and to differentiate induced pluripotent stem cells (iPSCs). The differentiation ability of iPSCs to neurons was justified by various immunological stains for protein fluorescence. The effect of PBCA NP/cmvNT-3-HRE complexes on treating ICH rats was studied by immunostaining, western blotting and Nissl staining. We found that the treatments with PBCA NP/cmvNT-3-HRE complexes increased the capability of differentiating iPSCs to express NT-3, TrkC and MAP-2. Moreover, PBCA NPs could protect cmvNT-3-HRE against degradation with EcoRI/PstI and DNase I in vitro and raise the delivery across the BBB in vivo. The administration of PBCA NP/cmvNT-3-HRE complexes increased the expression of NT-3, inhibited the expression of apoptosis-inducing factor, cleaved caspase-3 and DNA fragmentation, and reduced the cell death rate after ICH in vivo. PBCA NPs are demonstrated as an appropriate delivery system for carrying cmvNT-3-HRE to the brain for ICH therapy. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
48. Cloning and functional characterization of human Pak1 promoter by steroid hormones
- Author
-
Ganesh Venkatraman, Suresh K. Rayala, and Swetha Raghavan
- Subjects
0301 basic medicine ,Transcription, Genetic ,Response element ,Breast Neoplasms ,Biology ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Progesterone receptor ,Gene expression ,Genetics ,medicine ,Transcriptional regulation ,Humans ,Electrophoretic mobility shift assay ,Cloning, Molecular ,Promoter Regions, Genetic ,Progesterone ,Hormone response element ,Binding Sites ,Estradiol ,General Medicine ,Up-Regulation ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,p21-Activated Kinases ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Female ,Carcinogenesis ,Chromatin immunoprecipitation - Abstract
P21-activated kinase 1 (Pak1) is known to be involved in a plethora of functions including cell growth, survival and can lead to cell transformation and tumor progression especially in breast tissue. Multiple studies have shown Pak1 dysregulation as a change in DNA copy number as well as gene expression levels, suggesting many regulatory mechanisms at transcriptional and translational level. However, very little is known about the transcriptional regulation of the human Pak1 promoter. Here, we focus on Pak1 promoter regulation by steroid hormones along with their respective receptors that are also crucial players in breast tissue function and tumorigenesis. Our results show high Pak1 expression in breast cancer cell lines and in breast tumor tissue. It also suggests that Pak1 is hormone responsive, whose expression can be modulated by steroid hormones namely, estrogen in the form of 17β-estradiol (E2) and progesterone (P4). Sequence analysis of a 3.2kb Pak1 proximal promoter region shows the presence of PRE (progesterone response element) and ERE (estrogen response element) half sites, that were further cloned and characterized. Results from promoter analysis showed that Pak1 promoter activity is mediated by PR via its binding to PRE present on the Pak1 promoter that was further reaffirmed in vitro by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation assay (ChIP). Our results together suggest that it is the PR isoform B regulates Pak1 promoter. To our knowledge, this is the first study to report the detailed characterization and transcriptional regulation of the human Pak1 promoter by steroid hormones.
- Published
- 2018
49. Genome-wide mapping of estrogen receptor α binding sites by ChIP-seq to identify genes related to sexual maturity in hens
- Author
-
Yuxia Chen, Zhenjie Yuan, Yunliang Jiang, Yi Li, Miao Guo, and Xiaoli Guo
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,medicine.drug_class ,Estrogen receptor ,Biology ,Response Elements ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Gene Regulatory Networks ,Protein Interaction Maps ,Binding site ,Gene ,Hormone response element ,Binding Sites ,Ovary ,Estrogen Receptor alpha ,Gene Expression Regulation, Developmental ,DNA ,General Medicine ,Sex Determination Processes ,Cell biology ,030104 developmental biology ,Estrogen ,030220 oncology & carcinogenesis ,biology.protein ,Female ,CREB1 ,Chickens ,Chromatin immunoprecipitation ,Estrogen receptor alpha - Abstract
In ovarian follicle development, estrogen acts as a regulatory molecule to mediate proliferation and differentiation of follicular cells. ERα (estrogen receptor α) exerts regulatory function classically by binding directly to the estrogen response element, recruiting co-factors and activating or repressing transcription in response to E2. In this study, we used ChIP-seq to map ERα-binding sites in ovaries of Hy-line Brown commercial hens at 45d, 90d and 160d. In total, 24,886, 21,680 and 23,348 binding sites were identified in the ovaries of hens at 45d, 90d and 160d, which are linked to 86, 83 and 74 genes, respectively. The PPI network contains 47 protein nodes and 164 interaction edges, among which, AKT1 (V-Akt Murine Thymoma Viral Oncogene Homolog 1) and ACTN2 (Actinin Alpha 2) with the highest weight in the network, followed by CREB1 (CAMP Responsive Element Binding Protein 1), and EPHA5 (EPH Receptor A5) were identified. These genes are likely related to sexual maturity in hens. This study also provides insight into the regulation of the ERα target gene networks and a reference for understanding ERα-regulated transcription.
- Published
- 2018
50. Genomic Analyses of Hormone Signaling and Gene Regulation.
- Author
-
Cheung, Edwin and Kraus, W. Lee
- Subjects
- *
GENE expression , *GENOMICS , *TRANSCRIPTION factors , *NUCLEAR receptors (Biochemistry) , *GENETIC regulation , *BIOINFORMATICS - Abstract
Many cellular signaling pathways ultimately control specific patterns of gene expression in the nucleus through a variety of signal-regulated transcription factors (TFs), including nuclear hormone receptors (NRs). The advent of genomic technologies for examining signal-regulated transcriptional responses and TF binding on a genomic scale has dramatically increased our understanding of the cellular programs that control hormonal signaling and gene regulation. Studies of TFs, especially NRs, using genomic approaches have revealed novel and unexpected features of hormone-regulated transcription, and a global view is beginning to emerge. In this review, we discuss the genomic methodologies that have been applied to the study of hormone-regulated gene expression, the results that have been obtained from using them, and the future prospects for these approaches. Given the wealth of information about hormone-dependent gene regulation by NRs, we have focused this review on the knowledge gained from genomic studies of their function. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.