151
|
Numakawa T, Odaka H, Adachi N, Chiba S, Ooshima Y, Matsuno H, Nakajima S, Yoshimura A, Fumimoto K, Hirai Y, Kunugi H. Basic fibroblast growth factor increased glucocorticoid receptors in cortical neurons through MAP kinase pathway. Neurochem Int 2018; 118:217-224. [PMID: 29958871 DOI: 10.1016/j.neuint.2018.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/30/2018] [Accepted: 06/25/2018] [Indexed: 01/09/2023]
Abstract
Prolonged and intense stress chronically increases blood concentration of glucocorticoids, which in turn causes downregulation of glucocorticoid receptor (GR) in the central nervous system (CNS). This process has been suggested to be involved in the pathogenesis of major depressive disorder (MDD). Here, we found that basic fibroblast growth factor (bFGF) increased the expression of GR in the rat cerebral cortex and cultured cortical neurons and restored the reduced GR expression caused by glucocorticoid exposure. Among intracellular signaling pathways stimulated by bFGF, extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathway was responsible for the upregulation of GR. The bFGF-induced GR was functional as a transcription factor to enhance transcription of a target gene. Because high stress augments bFGF levels in the brain, it is likely that bFGF plays a compensating role for reduced GR expression after stress and thus should be studied as a therapeutic target for the treatment of MDD.
Collapse
Affiliation(s)
- Tadahiro Numakawa
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Haruki Odaka
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Naoki Adachi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan.
| | - Shuichi Chiba
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Yoshiko Ooshima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Administrative Section of Radiation Protection, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Hitomi Matsuno
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| | - Shingo Nakajima
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Endowed Research Division of Human Welfare Sciences, Ochanomizu University, Tokyo, Japan
| | - Aya Yoshimura
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan; Education and Research Facility of Animal Models for Human Diseases, Center for Research Promotion and Support, Fujita Health University, Aichi, Japan
| | - Kazuhiro Fumimoto
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Yohei Hirai
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Gakuen 2-1, Sanda City, Hyogo, 669-1337, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, NCNP, Tokyo, Japan
| |
Collapse
|
152
|
Marczell I, Balogh P, Nyiro G, Kiss AL, Kovacs B, Bekesi G, Racz K, Patocs A. Membrane-bound estrogen receptor alpha initiated signaling is dynamin dependent in breast cancer cells. Eur J Med Res 2018; 23:31. [PMID: 29880033 PMCID: PMC5992704 DOI: 10.1186/s40001-018-0328-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 05/19/2018] [Indexed: 01/22/2023] Open
Abstract
Background Although membrane-associated estrogen receptors (mERs) have been known to play important role in steroid-induced signal transmission, we still know little about their function in the estrogen-induced proliferation of breast cancer cells. Methods In our current work we tried to separate membrane-initiated estrogen receptor signaling from the overall estrogenic effect in MCF-7 breast carcinoma cells. Re-analyzing expression data from multiple microarray experiments, we selected a set of key regulatory genes involved in proliferation regulation and estrogen signaling to monitor estrogen-induced transcription changes. We then compared these expression changes after 17β-estradiol and a membrane receptor selective estrogen–BSA treatment using quantitative real-time PCR. In order to follow receptor trafficking we used light and electron microscopy. Results Our quantitative real-time PCR results confirmed that the selective membrane receptor agonist, estrogen–BSA induces similarly pronounced expression changes regarding these genes as 17β-estradiol. Morphological study revealed that the membrane-bound form of classical estrogen receptor alpha is internalized after ligand binding via dynamin-dependent, caveola-mediated endocytosis. Inhibition of this internalization with dynamin inhibitor, dynasore practically abolished the regulatory effect of E2-BSA, suggesting that interaction and internalization with the scaffold protein is necessary for effective signaling. Conclusions The physiological role of plasma membrane estrogen receptor alpha is intensively studied, yet there are still several aspects of it to be resolved. The dynamin-dependent, ligand-mediated internalization of mERs seems to play an important role in estrogen signaling. Our results may serve as another example of how membrane initiated estrogen signaling and nuclear receptor initiated signaling overlap and form an intertwined system. Electronic supplementary material The online version of this article (10.1186/s40001-018-0328-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Istvan Marczell
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary
| | - Petra Balogh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Gabor Nyiro
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary.,Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, Szentkirályi str. 46., 1088, Hungary
| | - Anna L Kiss
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Balazs Kovacs
- Department of Aquaculture, Szent Istvan University, Godollo, Hungary
| | - Gabor Bekesi
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary
| | - Karoly Racz
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary.,Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Attila Patocs
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary. .,HAS-SE 'Lendület' Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, 46. Szentkiralyi str, 1088, Hungary. .,Department of Laboratory Medicine, Semmelweis University, Budapest, Nagyvárad sq 4, 1089, Hungary.
| |
Collapse
|
153
|
Pollard KJ, Wartman HD, Daniel JM. Previous estradiol treatment in ovariectomized mice provides lasting enhancement of memory and brain estrogen receptor activity. Horm Behav 2018; 102:76-84. [PMID: 29742445 PMCID: PMC6004337 DOI: 10.1016/j.yhbeh.2018.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/18/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022]
Affiliation(s)
| | | | - Jill M Daniel
- Tulane University, Tulane Brain Institute, United States; Tulane University, Department of Psychology, United States
| |
Collapse
|
154
|
Zearalenone Promotes Cell Proliferation or Causes Cell Death? Toxins (Basel) 2018; 10:toxins10050184. [PMID: 29724047 PMCID: PMC5983240 DOI: 10.3390/toxins10050184] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 04/26/2018] [Accepted: 04/27/2018] [Indexed: 02/06/2023] Open
Abstract
Zearalenone (ZEA), one of the mycotoxins, exerts different mechanisms of toxicity in different cell types at different doses. It can not only stimulate cell proliferation but also inhibit cell viability, induce cell apoptosis, and cause cell death. Thus, the objective of this review is to summarize the available mechanisms and current evidence of what is known about the cell proliferation or cell death induced by ZEA. An increasing number of studies have suggested that ZEA promoted cell proliferation attributing to its estrogen-like effects and carcinogenic properties. What’s more, many studies have indicated that ZEA caused cell death via affecting the distribution of the cell cycle, stimulating oxidative stress and inducing apoptosis. In addition, several studies have revealed that autophagy and some antioxidants can reverse the damage or cell death induced by ZEA. This review thoroughly summarized the metabolic process of ZEA and the molecular mechanisms of ZEA stimulating cell proliferation and cell death. It concluded that a low dose of ZEA can exert estrogen-like effects and carcinogenic properties, which can stimulate the proliferation of cells. While, in addition, a high dose of ZEA can cause cell death through inducing cell cycle arrest, oxidative stress, DNA damage, mitochondrial damage, and apoptosis.
Collapse
|
155
|
Blaschka C, Schuler G, Sánchez-Guijo A, Zimmer B, Feller S, Kotarski F, Wudy SA, Wrenzycki C. Occurrence of sulfonated steroids and ovarian expression of steroid sulfatase and SULT1E1 in cyclic cows. J Steroid Biochem Mol Biol 2018; 179:79-87. [PMID: 29262378 DOI: 10.1016/j.jsbmb.2017.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 12/28/2022]
Abstract
Historically sulfonated steroids were primarily considered as inactive metabolites destined for elimination. However, more recently they have been increasingly recognized as precursors for the production of bioactive steroids in target tissues and as functional molecules without preceding hydrolysis. In order to comprehensively characterize their occurrence in cyclic cows and their formation and hydrolysis in bovine ovarian steroidogenesis, ovaries from cyclic cows were screened for the expression of oestrogen sulfotransferase (SULTE1) and steroid sulfatase (STS) by Western blot and immunohistochemistry. Moreover, a broad spectrum of 13 sulfonated steroids was measured applying liquid chromatography-tandem mass spectrometry (LC-MS/MS) in blood samples collected from three cycling heifers during defined stages of the ovarian cycle and in fluid obtained from ovarian follicles of different size. SULT1E1 was undetectable in ovarian tissues. For STS only a weak immunostaining was found predominantly in granulosa cells of larger follicles. However, no specific band occurred in Western blot. In blood, concentrations of all sulfonated steroids investigated were below the limit of quantification (LOQ). In follicular fluid, only cholesterol sulfate was measured in considerable concentrations (328.3 ± 63.8 ng/ml). However, the role of cholesterol sulfate in bovine follicular steroidogenesis remains unclear as concentrations were obviously unrelated to follicular size. The remaining sulfonated steroids investigated were undetectable or only slightly exceeded LOQ in a minor proportion of samples. The results are clearly contrary to a role of sulfonated steroids as important precursors, intermediates or products of bovine ovarian steroidogenesis.
Collapse
Affiliation(s)
- Carina Blaschka
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Gerhard Schuler
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Alberto Sánchez-Guijo
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstrasse 10-12, D-35392 Giessen, Germany
| | - Bettina Zimmer
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Sabine Feller
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Franziska Kotarski
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany
| | - Stefan A Wudy
- Steroid Research and Mass Spectrometry Unit, Center of Child and Adolescent Medicine, Faculty of Medicine, Justus-Liebig-University Giessen, Feulgenstrasse 10-12, D-35392 Giessen, Germany
| | - Christine Wrenzycki
- Clinic for Veterinary Obstetrics, Gynaecology and Andrology, Department of Molecular Reproductive Medicine, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Frankfurter Strasse 106, D-35392 Giessen, Germany.
| |
Collapse
|
156
|
Zhang H, Song T, Yang Y, Fu C, Li J. Exploring the Interaction Mechanism Between Cyclopeptide DC3 and Androgen Receptor Using Molecular Dynamics Simulations and Free Energy Calculations. Front Chem 2018; 6:119. [PMID: 29755968 PMCID: PMC5932393 DOI: 10.3389/fchem.2018.00119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/30/2018] [Indexed: 01/10/2023] Open
Abstract
Androgen receptor (AR) is a key target in the discovery of anti-PCa (Prostate Cancer) drugs. Recently, a novel cyclopeptide Diffusa Cyclotide-3 (DC3), isolated from Hedyotisdiffusa, has been experimentally demonstrated to inhibit the survival and growth of LNCap cells, which typically express T877A-mutated AR, the most frequently detected point mutation of AR in castration-resistant prostate cancer (CRPC). But the interaction mechanism between DC3 and AR is not clear. Here in this study we aim to explore the possible binding mode of DC3 to T877A-mutated AR from molecular perspective. Firstly, homology modeling was employed to construct the three-dimensional structure of the cyclopeptide DC3 using 2kux.1.A as the template. Then molecular docking, molecular dynamics (MD) simulations, and molecular mechanics/generalized Born surface area (MM-GBSA) methods were performed to determine the bind site and explore the detailed interaction mechanism of DC3-AR complex. The obtained results suggested that the site formed by H11, loop888-893, and H12 (site 2) was the most possible position of DC3 binding to AR. Besides, hydrogen bonds, hydrophobic, and electrostatic interactions play dominant roles in the recognition and combination of DC3-AR complex. The essential residues dominant in each interaction were specifically revealed. This work facilitates our understanding of the interaction mechanism of DC3 binding to AR at the molecular level and contributes to the rational cyclopeptide drug design for prostate cancer.
Collapse
Affiliation(s)
- Huimin Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Tianqing Song
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Yizhao Yang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, China
| | - Chenggong Fu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Jiazhong Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
157
|
Abstract
The Androgen Receptor (AR), a member of the steroid hormone receptor family, plays important roles in the physiology and pathology of diverse tissues. AR ligands, which include circulating testosterone and locally synthesized dihydrotestosterone, bind to and activate the AR to elicit their effects. Ubiquitous expression of the AR, metabolism and cross reactivity with other receptors limit broad therapeutic utilization of steroidal androgens. However, the discovery of selective androgen receptor modulators (SARMs) and other tissue-selective nuclear hormone receptor modulators that activate their cognate receptors in a tissue-selective manner provides an opportunity to promote the beneficial effects of androgens and other hormones in target tissues with greatly reduced unwanted side-effects. In the last two decades, significant resources have been dedicated to the discovery and biological characterization of SARMs in an effort to harness the untapped potential of the AR. SARMs have been proposed as treatments of choice for various diseases, including muscle-wasting, breast cancer, and osteoporosis. This review provides insight into the evolution of SARMs from proof-of-concept agents to the cusp of therapeutic use in less than two decades, while covering contemporary views of their mechanisms of action and therapeutic benefits.
Collapse
Affiliation(s)
- Ramesh Narayanan
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | | | - James T Dalton
- College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
158
|
Salkho NM, Paul V, Kawak P, Vitor RF, Martins AM, Al Sayah M, Husseini GA. Ultrasonically controlled estrone-modified liposomes for estrogen-positive breast cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:462-472. [DOI: 10.1080/21691401.2018.1459634] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Najla M. Salkho
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Pierre Kawak
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Rute F. Vitor
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ana M. Martins
- California Institute for Quantitative Biosciences, Berkeley, CA, USA
| | - Mohammad Al Sayah
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
159
|
Frick KM, Tuscher JJ, Koss WA, Kim J, Taxier LR. Estrogenic regulation of memory consolidation: A look beyond the hippocampus, ovaries, and females. Physiol Behav 2018; 187:57-66. [PMID: 28755863 PMCID: PMC5787049 DOI: 10.1016/j.physbeh.2017.07.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/14/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022]
Abstract
The potent estrogen 17β-estradiol (E2) has long been known to regulate the hippocampus and hippocampal-dependent memories in females, and research from the past decade has begun to shed light on the molecular mechanisms through which E2 mediates memory formation in females. Although E2 can also regulate hippocampal function in males, relatively little is known about how E2 influences memory formation in males, or whether sex differences in underlying mechanisms exist. This review, based on a talk given in April 2017 at the American University symposium entitled, "Sex Differences: From Neuroscience to the Clinic and Beyond", first provides an overview of the molecular mechanisms in the dorsal hippocampus through which E2 enhances memory consolidation in ovariectomized female mice. Next, newer research is described demonstrating key roles for the prefrontal cortex and de novo hippocampal E2 synthesis to the memory-enhancing effects of E2 in females. The review then discusses the effects of de novo and exogenous E2 on hippocampal memory consolidation in both sexes, and putative sex differences in the underlying molecular mechanisms through which E2 enhances memory formation. The review concludes by discussing the importance and implications of sex differences in the molecular mechanisms underlying E2-induced memory consolidation for human health.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States.
| | - Jennifer J Tuscher
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Wendy A Koss
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Jaekyoon Kim
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| | - Lisa R Taxier
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, United States
| |
Collapse
|
160
|
Cottone E, Orso F, Biglia N, Sismondi P, De Bortoli M. Role of Coactivators and Corepressors in Steroid and Nuclear Receptor Signaling: Potential Markers of Tumor Growth and Drug Sensitivity. Int J Biol Markers 2018; 16:151-66. [PMID: 11605727 DOI: 10.1177/172460080101600301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Nuclear receptors regulate target gene expression in response to steroid and thyroid hormones, retinoids, vitamin D and other ligands. These ligand-dependent transcription factors function by contacting various nuclear cooperating proteins, called coactivators and corepressors, which mediate local chromatin remodeling as well as communication with the basal transcriptional apparatus. Nuclear receptors and their coregulatory proteins play a role in cancer and other diseases, one leading example being the estrogen receptor pathway in breast cancer. Coregulators are often present in limiting amounts in cell nuclei and modifications of their level of expression and/or structure lead to alterations in nuclear receptor functioning, which may be as pronounced as a complete inversion of signaling, i.e. from stimulating to repressing certain genes in response to an identical stimulus. In addition, hemizygous knock-out of certain coactivator genes has been demonstrated to produce cancer-prone phenotypes in mice. Thus, assessment of coactivator and corepressor expression and structure in tumors may turn out to be essential to determine the role of nuclear receptors in cancer and to predict prognosis and response to therapy.
Collapse
Affiliation(s)
- E Cottone
- Department of Animal and Human Biology, University of Turin, Italy
| | | | | | | | | |
Collapse
|
161
|
Bowen L, Miles AK, Waters S, Gustine D, Joly K, Hilderbrand G. Using Gene Transcription to Assess Ecological and Anthropological Stressors in Brown Bears. ECOHEALTH 2018; 15:121-131. [PMID: 29168050 DOI: 10.1007/s10393-017-1287-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/25/2017] [Accepted: 10/30/2017] [Indexed: 06/07/2023]
Abstract
Increasingly, population- and ecosystem-level health assessments are performed using sophisticated molecular tools. Advances in molecular technology enable the identification of synergistic effects of multiple stressors on the individual physiology of different species. Brown bears (Ursus arctos) are an apex predator; thus, they are ideal candidates for detecting potentially ecosystem-level systemic perturbations using molecular-based tools. We used gene transcription to analyze 130 brown bear samples from three National Parks and Preserves in Alaska. Although the populations we studied are apparently stable in abundance and exist within protected and intact environments, differences in transcript profiles were noted. The most prevalent differences were among locations. The transcript patterns among groups reflect the influence of environmental factors, such as nutritional status, disease, and xenobiotic exposure. However, these profiles also likely represent baselines for each unique environment by which future measures can be made to identify early indication of population-level changes due to, for example, increasing Arctic temperatures. Some of those environmental changes are predicted to be potentially positive for brown bears, but other effects such as the manifestation of disease or indirect effects of oceanic acidification may produce negative impacts.
Collapse
Affiliation(s)
- Lizabeth Bowen
- U.S. Geological Survey, Western Ecological Research Center, University of California, Davis Field Station, Davis, CA, 95616, USA.
| | - A Keith Miles
- U.S. Geological Survey, Western Ecological Research Center, University of California, Davis Field Station, Davis, CA, 95616, USA
| | - Shannon Waters
- U.S. Geological Survey, Western Ecological Research Center, University of California, Davis Field Station, Davis, CA, 95616, USA
| | - Dave Gustine
- National Park Service, Grand Teton National Park, PO Box 170, Moose, WY, 83012, USA
| | - Kyle Joly
- National Park Service, Gates of the Arctic National Park and Preserve, 4175 Geist Road, Fairbanks, AK, 99709, USA
| | - Grant Hilderbrand
- U.S. Geological Survey, Alaska Science Center, Anchorage, AK, 99508, USA
| |
Collapse
|
162
|
Abstract
The nuclear receptor family of transcription factor proteins mediates endocrine function and plays critical roles in the development, physiology and pharmacology. Malfunctioning nuclear receptors are associated with several disease states. The functional activity of nuclear receptors is regulated by small molecular hormonal and synthetic molecules. Multiple sources of evidence have identified and distinguished between the different allosteric pathways initiated by ligands, DNA and cofactors such as co-activators and co-repressors. Also, these biophysical studies are attempting to determine how these pathways that regulate co-activator and DNA recognition can control gene transcription. Thus, there is a growing interest in determining the genome-scale impact of allostery in nuclear receptors. Today, it is accepted that a detailed understanding of the allosteric regulatory pathways within the nuclear receptor molecular complex will enable the development of efficient drug therapies in the long term.
Collapse
Affiliation(s)
- Elias J Fernandez
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, USA.
| |
Collapse
|
163
|
Balthazart J, Choleris E, Remage-Healey L. Steroids and the brain: 50years of research, conceptual shifts and the ascent of non-classical and membrane-initiated actions. Horm Behav 2018; 99:1-8. [PMID: 29305886 PMCID: PMC5880709 DOI: 10.1016/j.yhbeh.2018.01.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 12/30/2017] [Accepted: 01/02/2018] [Indexed: 11/22/2022]
Abstract
This brief commentary reviews key steps in the history of steroid endocrinology that have resulted in important conceptual shifts. Our understanding of the "Fast Effects of Steroids" now reflect substantial progress, including the major concept that steroids act rapidly on a variety of physiological and behavioral responses, via mechanisms that are too fast to be fully accounted for by classical receptor-dependent regulation of gene transcription. Several so-called 'non-classical' mechanisms have been identified and include binding to membrane receptors and regulating non genomic signaling cascades. We survey the discovery of steroids, the initial characterization of their intracellular receptors, key progress in the understanding of the genomic effects of steroids and then the progressive discovery of the rapid non-classical and membrane-initiated actions of steroids. Foundational discoveries about brain steroid synthesis in neural processes and terminals has converged with emerging evidence for the rapid actions of steroids on brain and behavior. Had the rapid effects of steroids in the central nervous system been discovered first, these molecules would likely now be considered as a class of neurotransmitter.
Collapse
Affiliation(s)
| | - Elena Choleris
- Department of Psychology and Neuroscience Program, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Luke Remage-Healey
- Center for Neuroendocrine Studies, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
164
|
Wang X, Zhang Q, Zhou Z, Liu M, Chen Y, Li J, Xu L, Guo J, Li Q, Yang J, Wang S. Retinoic acid receptor β, a potential therapeutic target in the inhibition of adenovirus replication. Antiviral Res 2018; 152:84-93. [PMID: 29421320 DOI: 10.1016/j.antiviral.2018.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
Abstract
Human adenoviruses (HAdVs) usually cause mild respiratory infections, but they can also lead to fatal outcomes for immunosuppressive patients. Unfortunately, there has been no specific anti-HAdV drug approved for medical use. A better understanding of the nature of virus-host interactions during infection is beneficial to the discovery of potential antiviral targets and new antiviral drugs. In this study, a time-course transcriptome analysis of HAdV-infected human lung epithelial cells (A549 cells) was performed to investigate virus-host interactions, and several key host molecules involved in the HAdV infection process were identified. The RARβ (retinoic acid receptor β) molecule, one of the upstream regulatory factors of differentially expressed genes (DEGs), played important roles in HAdV replication. The results of reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting showed that RARβ mRNA and protein were downregulated by HAdV infection in the A549 cells. The knockdown of RARβ by RARβ siRNA increased the HAdV production and the overexpression of RARβ decreased the HAdV production. Furthermore, FDA-approved Tazarotene, which is an RAR selective agonist with relatively more selectivity for RARβ, was found to inhibit HAdV replication in vitro. Taken together, our study presents a key host molecule in adenovirus infection, which could be developed as a potential host target to an anti-adenovirus drug. In addition, this study provides evidence for the re-exploitation of an FDA-approved small molecule for therapeutic applications in adenovirus replication.
Collapse
Affiliation(s)
- Xiaolong Wang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Qiling Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Zhe Zhou
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Manjiao Liu
- Beijing Computing Center, Beijing Academy of of Science and Technology, Beijing 100850, PR China; The Key Laboratory of Beijing Cloud Computing Technology and Application, Beijing 100850, PR China
| | - Yubao Chen
- Beijing Computing Center, Beijing Academy of of Science and Technology, Beijing 100850, PR China; The Key Laboratory of Beijing Cloud Computing Technology and Application, Beijing 100850, PR China
| | - Jianbo Li
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Linlin Xu
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Jing Guo
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China
| | - Qingjun Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jing Yang
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| | - Shengqi Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, PR China.
| |
Collapse
|
165
|
Sakane Y, Iida M, Hasebe T, Fujii S, Buchholz DR, Ishizuya-Oka A, Yamamoto T, Suzuki KIT. Functional analysis of thyroid hormone receptor beta in Xenopus tropicalis founders using CRISPR-Cas. Biol Open 2018; 7:bio.030338. [PMID: 29358165 PMCID: PMC5829506 DOI: 10.1242/bio.030338] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Amphibians provide an ideal model to study the actions of thyroid hormone (TH) in animal development because TH signaling via two TH receptors, TRα and TRβ, is indispensable for amphibian metamorphosis. However, specific roles for the TRβ isoform in metamorphosis are poorly understood. To address this issue, we generated trβ-disrupted Xenopus tropicalis tadpoles using the CRISPR-Cas system. We first established a highly efficient and rapid workflow for gene disruption in the founder generation (F0) by injecting sgRNA and Cas9 ribonucleoprotein. Most embryos showed severe mutant phenotypes carrying high somatic mutation rates. Utilizing this founder analysis system, we examined the role of trβ in metamorphosis. trβ-disrupted pre-metamorphic tadpoles exhibited mixed responsiveness to exogenous TH. Specifically, gill resorption and activation of several TH-response genes, including trβ itself and two protease genes, were impaired. However, hind limb outgrowth and induction of the TH-response genes, klf9 and fra-2, were not affected by loss of trβ Surprisingly, trβ-disrupted tadpoles were able to undergo spontaneous metamorphosis normally, except for a slight delay in tail resorption. These results indicate TRβ is not required but contributes to the timing of resorptive events of metamorphosis.
Collapse
Affiliation(s)
- Yuto Sakane
- Department of Mathematical and Life Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Midori Iida
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Takashi Hasebe
- Department of Biology, Nippon Medical School, Musashino, Tokyo 180-0023, Japan
| | - Satoshi Fujii
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan
| | - Daniel R Buchholz
- Department of Biological Sciences, University of Cincinnati, 312 Clifton Ct., Cincinnati, OH, 45221, USA
| | - Atsuko Ishizuya-Oka
- Department of Biology, Nippon Medical School, Musashino, Tokyo 180-0023, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Ken-Ichi T Suzuki
- Department of Mathematical and Life Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| |
Collapse
|
166
|
Abstract
The amphibian Xenopus laevis has long been used as a model for studying vertebrate cell and developmental biology largely due to the easiness to manipulate this system in vivo and in vitro. While most of the developmental studies have been on Xenopus embryogenesis, considerable efforts have been made to understand its metamorphosis, a process mimicking postembryonic development in mammals when many organs mature into their adult forms in the presence of high levels of thyroid hormone (T3). Amphibian metamorphosis is totally dependent on T3 and offers a number of advantages for experimental analyses compared to the late stage, uterus-enclosed mammalian embryos. Earlier studies on metamorphosis in Xenopus laevis have revealed dual functions of T3 receptors (TR) during premetamorphic development and metamorphosis as well as important roles of TR-interacting corepressors and coactivators during these two periods, respectively. The development of gene-editing technologies that functions in amphibians in recent years has made possible for the first time to study function of endogenous TRs, especially in the highly related diploid anuran species Xenopus tropicalis. Here, we first review the current mechanistic understanding of the regulation of metamorphosis by T3 and TR, and then describe a detailed method to use TALEN to knock out TRα for studying its role in gene regulation by T3 in vivo and Xenopus development.
Collapse
Affiliation(s)
- Liezhen Fu
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Luan Wen
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yun-Bo Shi
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
167
|
Ameen GI, Mora S. Cbl downregulation increases RBP4 expression in adipocytes of female mice. J Endocrinol 2018; 236:29-41. [PMID: 29114012 PMCID: PMC5744582 DOI: 10.1530/joe-17-0359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 11/07/2017] [Indexed: 11/30/2022]
Abstract
Obesity leads to adipose tissue dysfunction, insulin resistance and diabetes. Adipose tissue produces adipokines that contribute to regulate insulin sensitivity. In turn, insulin stimulates the production and release of some adipokines. Casitas-b-lymphoma proteins (c-Cbl, Cbl-b and Cbl3) are intracellular adaptor signalling proteins that are rapidly phosphorylated by activation of tyrosine kinase receptors. c-Cbl is rapidly phosphorylated by insulin in adipocytes. Here, we tested the hypothesis that Cbl signalling regulates adipokine expression in adipose tissue. We determined the adipokine profile of WAT of Cbl-/- and Cbl+/+ mice in the C57BL6 background. Female Cbl-/- mice exhibited altered expression of adiponectin, leptin and RBP4 in visceral adipose tissue, while no significant changes were seen in male mice. TNFα and IL6 levels were unaffected by Cbl depletion. RBP4 expression was unchanged in liver. Adipose tissue of Cbl-/- animals showed increased basal activation of extracellular regulated kinases (ERK1/2) compared to Cbl+/+. c-Cbl knockdown in 3T3L1 adipocytes also increased basal ERK phosphorylation and RBP4 expression. Inhibition of ERK1/2 phosphorylation in Cbl-depleted 3T3L1 adipocytes or in adipose tissue explants of Cbl-/- mice reduced RBP4 mRNA. 17β-Estradiol increased RBP4 mRNA in adipocytes. Cbl depletion did not change ER expression but increased phosphorylation of ERα at S118, a target site for ERK1/2. ERK1/2 inhibition reduced phosphoER and RBP4 levels. These findings suggest that Cbl contributes to regulate RBP4 expression in adipose of female mice through ERK1/2-mediated activation of ERα. Since Cbl signalling is compromised in diabetes, these data highlight a novel mechanism that upregulates RBP4 locally.
Collapse
Affiliation(s)
- Gulizar Issa Ameen
- Department of Cellular and Molecular PhysiologyInstitute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Silvia Mora
- Department of Cellular and Molecular PhysiologyInstitute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
168
|
Padilla Colón CJ, Molina-Vicenty IL, Frontera-Rodríguez M, García-Ferré A, Rivera BP, Cintrón-Vélez G, Frontera-Rodríguez S. Muscle and Bone Mass Loss in the Elderly Population: Advances in diagnosis and treatment. JOURNAL OF BIOMEDICINE (SYDNEY, NSW) 2018; 3:40-49. [PMID: 30505650 PMCID: PMC6261527 DOI: 10.7150/jbm.23390] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aging is the result of different functional changes leading to a substantial reduction of all human capabilities. A variety of anatomical and physiological changes occur with advancing age. These changes are more evident in the elderly population. There are various methods to measure muscle and bone mass loss, but the dual X-ray absorptiometry (DXA) is considered one of the most efficient. The elderly population (65 years and older) has been increasing throughout the years. Loss of muscle mass (sarcopenia) and loss bone mass (osteopenia or osteoporosis) with advancing age, when untreated, represent a major public health problem for the elderly population and may result in loss of independence in later life. Untreated age-related sarcopenia and osteopenia/osteoporosis increase the risk for falls and fractures, making older individuals more susceptible to the development of mobility limitations or severe disabilities that ultimately affect their capacity for independence. In this review, we will discuss the muscle and bone mass loss in the elderly population and advances in diagnosis and treatment.
Collapse
Affiliation(s)
- Carlos J. Padilla Colón
- Department of Education, Physical Education and Health Programs, San Juan, PR, USA
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
| | - Irma L. Molina-Vicenty
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
- Department of Radiological Sciences, University of Puerto Rico, Medical Sciences Campus, San Juan, PR, USA
| | - María Frontera-Rodríguez
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Alejandra García-Ferré
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | | | - Gerardo Cintrón-Vélez
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Sebastián Frontera-Rodríguez
- Research and Development Service (151), VA Caribbean Healthcare System, San Juan PR, USA
- Department of Biology, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| |
Collapse
|
169
|
Mondal S, Mugesh G. Novel thyroid hormone analogues, enzyme inhibitors and mimetics, and their action. Mol Cell Endocrinol 2017; 458:91-104. [PMID: 28408161 DOI: 10.1016/j.mce.2017.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/07/2017] [Accepted: 04/07/2017] [Indexed: 12/31/2022]
Abstract
Thyroid hormones (THs) play key roles in modulating the overall metabolism of the body, protein synthesis, fat metabolism, neuronal and bone growth, and cardiovascular as well as renal functions. In this review, we discuss on the thyroid hormone synthesis and activation, thyroid hormone receptors (TRs) and mechanism of action, applications of thyroid hormone analogues, particularly the compounds that are selective ligands for TRβ receptors, or enzyme inhibitors for the treatment of thyroidal disorders with a specific focus on thyroid peroxidase and iodothyronine deiodinases. We also discuss on the development of small-molecule deiodinase mimetics and their mechanism of deiodination, as these compounds have the potential to regulate the thyroid hormone levels.
Collapse
Affiliation(s)
- Santanu Mondal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India
| | - Govindasamy Mugesh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
170
|
Ezzati M, Roshangar L, Soleimani Rad J, Karimian N. Evaluating The Effect of Melatonin on HAS2, and PGR expression, as well as Cumulus Expansion, and Fertility Potential in Mice. CELL JOURNAL 2017; 20:108-112. [PMID: 29308626 PMCID: PMC5759672 DOI: 10.22074/cellj.2018.4894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/18/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Infertility is a worldwide health problem which affects approximately 15% of sexually active couples. One of the factors influencing the fertility is melatonin. Also, protection of oocytes and embryos from oxidative stress inducing chemicals in the culture medium is important. The aim of the present study was to investigate if melatonin could regulate hyaluronan synthase-2 (HAS2) and Progesterone receptor (PGR) expressions in the cumulus cells of mice oocytes and provide an in vitro fertilization (IVF) approach. MATERIALS AND METHODS In this experimental study, for this purpose, 30 adult female mice and 15 adult male mice were used. The female mice were superovulated using 10 U of pregnant mare serum gonadotropin (PMSG) and 24 hours later, 10 U of human chorionic gonadotropin (hCG) were injected. Next, cumulus oocyte complexes (COCs) were collected from the oviducts of the female mice by using a matrix-flushing method. The cumulus cells were cultured with melatonin 10 μM for 6 hours and for real-time reverse transcription-polymerase chain reaction (RT-PCR) was used for evaluation of HAS2 and PGR expression levels. The fertilization rate was evaluated through IVF. All the data were analyzed using a t test. RESULTS The results of this study showed that HAS2 and PGR expressions in the cumulus cells of the mice receiving melatonin increased in comparison to the control groups. Also, IVF results revealed an enhancement in fertilization rate in the experimental groups compared to the control groups. CONCLUSIONS To improve the oocyte quality and provide new approaches for infertility treatment, administration of melatonin as an antioxidant, showed promising results. Thus, it is concluded that fertility outcomes can be improved by melatonin it enhances PGR.
Collapse
Affiliation(s)
- Maryam Ezzati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Jafar Soleimani Rad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Karimian
- Department of Advanced Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
171
|
Yang Y, Leonard M, Zhang Y, Zhao D, Mahmoud C, Khan S, Wang J, Lower EE, Zhang X. HER2-Driven Breast Tumorigenesis Relies upon Interactions of the Estrogen Receptor with Coactivator MED1. Cancer Res 2017; 78:422-435. [PMID: 29187405 DOI: 10.1158/0008-5472.can-17-1533] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/18/2017] [Accepted: 11/20/2017] [Indexed: 12/21/2022]
Abstract
Studies of the estrogen receptor (ER) coactivator protein Mediator subunit 1 (MED1) have revealed its specific roles in pubertal mammary gland development and potential contributions to breast tumorigenesis, based on coamplification of MED1 and HER2 in certain breast cancers. In this study, we generated a mouse model of mammary tumorigenesis harboring the MMTV-HER2 oncogene and mutation of MED1 to evaluate its role in HER2-driven tumorigenesis. MED1 mutation in its ER-interacting LxxLL motifs was sufficient to delay tumor onset and to impair tumor growth, metastasis, and cancer stem-like cell formation in this model. Mechanistic investigations revealed that MED1 acted directly to regulate ER signaling through the downstream IGF1 pathway but not the AREG pathway. Our findings show that MED1 is critical for HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target.Significance: These findings identify an estrogen receptor-binding protein as a critical mediator of HER2-driven breast tumorigenesis, suggesting its candidacy as a disease-selective therapeutic target. Cancer Res; 78(2); 422-35. ©2017 AACR.
Collapse
Affiliation(s)
- Yongguang Yang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marissa Leonard
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio.,Graduate Program in Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yijuan Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Dan Zhao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Charif Mahmoud
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Shugufta Khan
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Elyse E Lower
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Xiaoting Zhang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio. .,Graduate Program in Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
172
|
Sánchez-Céspedes R, Fernández-Martínez MD, Raya A, Pineda C, López I, Millán Y. Vitamin D receptor expression in canine mammary gland and relationship with clinicopathological parameters and progesterone/oestrogen receptors. Vet Comp Oncol 2017; 16:E185-E193. [PMID: 29178579 DOI: 10.1111/vco.12371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023]
Abstract
The vitamin D receptor (VDR) belongs to the nuclear class II receptor family. VDR is a ligand transcription factor and mediates the actions of calcitriol, the active product of vitamin D synthesis. Nowadays, it is known that the biological actions of calcitriol include the capacity to modulate cancer features, such as proliferation and differentiation, apoptosis, angiogenesis, invasion and metastasis. VDR expression has been demonstrated in human breast cancer and vitamin D has emerged as a promising targeted therapy. We analyse the VDR expression in normal and neoplastic canine mammary tissue samples and its relationship with clinicopathological parameters and progesterone/oestrogens receptors (PR/ER). Expression of VDR, Ki67 (to evaluate the proliferation index, PI), PR and ER was assessed in 50 mammary gland tissue samples from 41 female dogs by immunohistochemistry. VDR-positive staining was found in the nuclei of both myoepithelial and luminal epithelial cell layers. VDR expression was higher in normal mammary tissue (37/37 cases, 100%) then followed by benign tumours (6/15 cases, 40%) and malignant tumours (9/34 cases, 26.5%) (P = .001). Female dogs aged ≥10 years had lower VDR expression compared with dogs younger (P = .017). Relationship between VDR and breed, number of tumours, tumour size, histologic subtype, histologic grade of malignancy, PI and PR and ER expression was not observed. Studies with more samples are necessary to further evaluate the possible role of VDR in the biological behaviour of canine mammary tumours, and to corroborate the possibility to use the dog as model for human breast cancer.
Collapse
Affiliation(s)
- R Sánchez-Céspedes
- Department of Comparative Pathology, University of Córdoba, Córdoba, Spain
| | | | - A Raya
- Department of Medicine and Animal Surgery, Veterinary Faculty, University of Córdoba, Córdoba, Spain
| | - C Pineda
- Department of Medicine and Animal Surgery, Veterinary Faculty, University of Córdoba, Córdoba, Spain
| | - I López
- Department of Medicine and Animal Surgery, Veterinary Faculty, University of Córdoba, Córdoba, Spain
| | - Y Millán
- Department of Comparative Pathology, University of Córdoba, Córdoba, Spain
| |
Collapse
|
173
|
de Bournonville C, Ball GF, Balthazart J, Cornil CA. Rapid changes in brain aromatase activity in the female quail brain following expression of sexual behaviour. J Neuroendocrinol 2017; 29. [PMID: 28990707 DOI: 10.1111/jne.12542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/14/2017] [Accepted: 10/02/2017] [Indexed: 12/29/2022]
Abstract
In male quail, oestrogens produced in the brain (neuro-oestrogens) exert a dual action on male sexual behaviour: they increase sexual motivation within minutes via mechanisms activated at the membrane but facilitate sexual performance by slower, presumably nuclear-initiated, mechanisms. Recent work indicates that neuro-oestrogens are also implicated in the control of female sexual motivation despite the presence of high circulating concentrations of oestrogens of ovarian origin. Interestingly, aromatase activity (AA) in the male brain is regulated in time domains corresponding to the slow "genomic" and faster "nongenomic" modes of action of oestrogens. Furthermore, rapid changes in brain AA are observed in males after sexual interactions with a female. In the present study, we investigated whether similar rapid changes in brain AA are observed in females allowed to interact sexually with males. A significant decrease in AA was observed in the medial preoptic nucleus after interactions that lasted 2, 5 or 10 minutes, although this decrease was no longer significant after 15 minutes of interaction. In the bed nucleus of the stria terminalis, a progressive decline of average AA was observed between 2 and 15 minutes, although it never reached statistical significance. AA in this nucleus was, however, negatively correlated with the sexual receptivity of the female. These data indicate that sexual interactions affect brain AA in females as in males in an anatomically specific manner and suggest that rapid changes in brain oestrogens production could also modulate female sexual behaviour.
Collapse
Affiliation(s)
- C de Bournonville
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - G F Ball
- Department of Psychology, University of Maryland, College Park, MD, USA
| | - J Balthazart
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium
| | - C A Cornil
- GIGA Neurosciences, Research Group in Behavioral Neuroendocrinology, University of Liège, Liège, Belgium
| |
Collapse
|
174
|
Bhatnagar S, Soni A, Kaushik S, Rikhi M, Santhoshkumar TR, Jayaram B. Nonsteroidal estrogen receptor isoform-selective biphenyls. Chem Biol Drug Des 2017; 91:620-630. [PMID: 29052968 DOI: 10.1111/cbdd.13126] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/18/2017] [Accepted: 10/04/2017] [Indexed: 12/17/2022]
Abstract
Estrogen receptor (ER) has been a therapeutic target to treat ER-positive breast cancer, most notably by agents known as selective estrogen receptor modulators (SERMs). However, resistance and severe adverse effects of known drugs gave impetus to the search for newer agents with better therapeutic profile. ERα and ERβ are two isoforms sharing 56% identity and having different physiological functions and expressions in various tissues. Only two residues differ in the active sites of the two isoforms motivating us to design isoform-selective ligands. Guided by computational docking and molecular dynamics simulations, we have designed, synthesized, and tested, substituted biphenyl-2,6-diethanones and their derivatives as potential agents targeting ERα. Four of the molecules synthesized exhibited preferential cytotoxicity in ERα+ cell line (MCF-7) compared to ERβ+ cell line (MDA-MB-231). Molecular dynamics (MD) in combination with molecular mechanics-generalized Born surface area (MM-GBSA) methods could account for binding selectivity. Further cotreatment and E-screen studies with known ER ligands-estradiol (E2 ) and tamoxifen (Tam)-indicated isoform-selective anti-estrogenicity in ERα+ cell line which might be ER-mediated. ERα siRNA silencing experiments further confirmed the ER selective nature of ligands.
Collapse
Affiliation(s)
- Seema Bhatnagar
- Amity Institute of Biotechnology, Amity University, Noida, India
| | - Anjali Soni
- Department of Chemistry and Supercomputing Facility for Bioinformatics and Computational Biology, Indian Institute of Technology, Hauz Khas, New Delhi, India
| | - Swati Kaushik
- Amity Institute of Biotechnology, Amity University, Noida, India.,Cancer Research Programme Lab1, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India
| | - Megha Rikhi
- Amity Institute of Biotechnology, Amity University, Noida, India
| | | | - Bhyravabhotla Jayaram
- Department of Chemistry and Supercomputing Facility for Bioinformatics and Computational Biology, Indian Institute of Technology, Hauz Khas, New Delhi, India.,Kusuma School of Biological Sciences, Indian Institute of Technology, Hauz Khas, New Delhi, India
| |
Collapse
|
175
|
Ranjan A, Ansari SA. Therapeutic potential of Mediator complex subunits in metabolic diseases. Biochimie 2017; 144:41-49. [PMID: 29061530 DOI: 10.1016/j.biochi.2017.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/16/2017] [Indexed: 01/16/2023]
Abstract
The multisubunit Mediator is an evolutionary conserved transcriptional coregulatory complex in eukaryotes. It is needed for the transcriptional regulation of gene expression in general as well as in a gene specific manner. Mediator complex subunits interact with different transcription factors as well as components of RNA Pol II transcription initiation complex and in doing so act as a bridge between gene specific transcription factors and general Pol II transcription machinery. Specific interaction of various Mediator subunits with nuclear receptors (NRs) and other transcription factors involved in metabolism has been reported in different studies. Evidences indicate that ligand-activated NRs recruit Mediator complex for RNA Pol II-dependent gene transcription. These NRs have been explored as therapeutic targets in different metabolic diseases; however, they show side-effects as targets due to their overlapping involvement in different signaling pathways. Here we discuss the interaction of various Mediator subunits with transcription factors involved in metabolism and whether specific interaction of these transcription factors with Mediator subunits could be potentially utilized as therapeutic strategy in a variety of metabolic diseases.
Collapse
Affiliation(s)
- Amol Ranjan
- Stowers Institute for Medical Research, 1000 E, 50th Street, Kansas City, MO, 64110, USA
| | - Suraiya A Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, UAE University, AlAin, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
176
|
Socha JK, Sechman A, Mika M, Hrabia A. Effect of growth hormone on steroid concentrations and mRNA expression of their receptor, and selected egg-specific protein genes in the chicken oviduct during pause in laying induced by fasting. Domest Anim Endocrinol 2017; 61:1-10. [PMID: 28551483 DOI: 10.1016/j.domaniend.2017.05.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 04/22/2017] [Accepted: 05/01/2017] [Indexed: 11/18/2022]
Abstract
This study was undertaken to examine the effect of growth hormone (GH) treatment during pause in laying on (1) the concentration of steroids in blood plasma and oviduct tissues, (2) the expression of mRNA of steroid receptors, and (3) the mRNA expression of selected egg-specific proteins in the chicken oviduct. A pause in egg laying was induced by food deprivation for 5 d, followed by feeding every other day, and then feeding daily from Day 10 onward. Birds were divided into three groups: control (n = 18) fed ad libitum, subjected to pause in laying (n = 18), and subjected to pause in laying and injected every day with 200 μg/kg BW of chicken GH (chGH; n = 18). The oviduct was isolated from hens of each group on Days 6 (when the oviduct was regressed), 13 (during oviduct recrudescence), and 17 or 20 (rejuvenated oviduct) of the experiment. Fasting caused a decrease in plasma concentrations of progesterone (P4), testosterone, and estradiol on Day 6 and a reduction in tissue concentrations of these steroids on Days 6 and 13. Fasting also caused an increased relative expression of estrogen receptor α and β (ERα, ERβ) and progesterone receptor (PR) in the magnum and shell gland on Day 6, increased ERα and PR in the magnum on Days 13 and 17 or 20, and increased androgen receptor (AR) mRNA in the magnum on Days 6 and 13 and in the shell gland on Day 13. A fasting-induced elevation in ovocalyxin-36 mRNA expression on Day 6 and a decrease in avidin mRNA on Days 6 and 13 and in ovocleidin-116 on Day 13 were also observed (P < 0.05 to P < 0.001). Administration of chGH abolished the fasting-induced decrease in the concentration of steroids in plasma and tissues. Furthermore, chGH enhanced the effect of fasting on mRNA expression of PR, ERα, and avidin in the magnum on Day 6, and ERα in the shell gland on Day 13. The gene expression of ovalbumin on Days 6 and 13, ovocalyxin-36 and ovocleidin-116 on Day 6 was decreased in chGH-treated chickens. In contrast, the expression of ovalbumin on Day 17 or 20 was increased (P < 0.05 to P < 0.001). The results obtained indicate that, by alterations in the concentration of steroid hormones and their receptor expression in the chicken oviduct, GH determines the rate of regression and rejuvenation of this organ during molting. Moreover, changes in the expression of selected egg proteins indicate that GH might be the regulator of the secretory activity of the hen oviduct.
Collapse
Affiliation(s)
- J K Socha
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland
| | - A Sechman
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland
| | - M Mika
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland
| | - A Hrabia
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland.
| |
Collapse
|
177
|
Kim DH, Kim HS, Hwang DS, Kim HJ, Hagiwara A, Lee JS, Jeong CB. Genome-wide identification of nuclear receptor (NR) genes and the evolutionary significance of the NR1O subfamily in the monogonont rotifer Brachionus spp. Gen Comp Endocrinol 2017; 252:219-225. [PMID: 28673513 DOI: 10.1016/j.ygcen.2017.06.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/09/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023]
Abstract
Nuclear receptors (NRs) are a large family of transcription factors that are involved in many fundamental biological processes. NRs are considered to have originated from a common ancestor, and are highly conserved throughout the whole animal taxa. Therefore, the genome-wide identification of NR genes in an animal taxon can provide insight into the evolutionary tendencies of NRs. Here, we identified all the NR genes in the monogonont rotifer Brachionus spp., which are considered an ecologically key species due to their abundance and world-wide distribution. The NR family was composed of 40, 32, 29, and 32 genes in the genomes of the rotifers B. calyciflorus, B. koreanus, B. plicatilis, and B. rotundiformis, respectively, which were classified into seven distinct subfamilies. The composition of each subfamily was highly conserved between species, except for NR1O genes, suggesting that they have undergone sporadic evolutionary processes for adaptation to their different environmental pressures. In addition, despite the dynamics of NR evolution, the significance of the conserved endocrine system, particularly for estrogen receptor (ER)-signaling, in rotifers was discussed on the basis of phylogenetic analyses. The results of this study may help provide a better understanding the evolution of NRs, and expand our knowledge of rotifer endocrine systems.
Collapse
Affiliation(s)
- Duck-Hyun Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hui-Su Kim
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Dae-Sik Hwang
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hee-Jin Kim
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - Atsushi Hagiwara
- Graduate School of Fisheries and Environmental Sciences, Nagasaki University, Nagasaki 852-8521, Japan
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| | - Chang-Bum Jeong
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
178
|
Enhancement of the bioavailability of a novel anticancer compound (acetyltanshinone IIA) by encapsulation within mPEG-PLGA nanoparticles: a study of formulation optimization, toxicity, and pharmacokinetics. Oncotarget 2017; 8:12013-12030. [PMID: 28061455 PMCID: PMC5355322 DOI: 10.18632/oncotarget.14481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 12/16/2016] [Indexed: 11/29/2022] Open
Abstract
The Poly (ethylene glycol) methyl ether-block-poly (lactide-co-glycolide) (mPEG-PLGA) nanoparticles carrying acetyltanshinone IIA (ATA), a novel anti-breast cancer agent, were prepared by ultrasonic emulsion method to enhance the bioavailability and reduce the toxicity. Systematic optimization of encapsulation process was achieved using an orthogonal design. Drug efficacy analysis showed that ATA nanoparticles were as effective as free ATA against estrogen receptor positive breast cancer cells, but much less toxic towards human endothelial cells. Furthermore, in zebrafish, ATA nanoparticles displayed much lower toxicity than free ATA. More importantly, the blood concentration of ATA nanoparticles indicated by 24 hour-area under the curve (AUC0-24h) was 10 times higher than free ATA. These results indicated the potential of ATA-loaded mPEG-PLGA nanoparticles for the delivery of ATA in a clinical formulation, and their potential for use in tumor therapy in the future.
Collapse
|
179
|
McDermott MSJ, Chumanevich AA, Lim CU, Liang J, Chen M, Altilia S, Oliver D, Rae JM, Shtutman M, Kiaris H, Győrffy B, Roninson IB, Broude EV. Inhibition of CDK8 mediator kinase suppresses estrogen dependent transcription and the growth of estrogen receptor positive breast cancer. Oncotarget 2017; 8:12558-12575. [PMID: 28147342 PMCID: PMC5355036 DOI: 10.18632/oncotarget.14894] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 12/15/2022] Open
Abstract
Hormone therapy targeting estrogen receptor (ER) is the principal treatment for ER-positive breast cancers. However, many cancers develop resistance to hormone therapy while retaining ER expression. Identifying new druggable mediators of ER function can help to increase the efficacy of ER-targeting drugs. Cyclin-dependent kinase 8 (CDK8) is a Mediator complex-associated transcriptional regulator with oncogenic activities. Expression of CDK8, its paralog CDK19 and their binding partner Cyclin C are negative prognostic markers in breast cancer. Meta-analysis of transcriptome databases revealed an inverse correlation between CDK8 and ERα expression, suggesting that CDK8 could be functionally associated with ER. We have found that CDK8 inhibition by CDK8/19-selective small-molecule kinase inhibitors, by shRNA knockdown or by CRISPR/CAS9 knockout suppresses estrogen-induced transcription in ER-positive breast cancer cells; this effect was exerted downstream of ER. Estrogen addition stimulated the binding of CDK8 to the ER-responsive GREB1 gene promoter and CDK8/19 inhibition reduced estrogen-stimulated association of an elongation-competent phosphorylated form of RNA Polymerase II with GREB1. CDK8/19 inhibitors abrogated the mitogenic effect of estrogen on ER-positive cells and potentiated the growth-inhibitory effects of ER antagonist fulvestrant. Treatment of estrogen-deprived ER-positive breast cancer cells with CDK8/19 inhibitors strongly impeded the development of estrogen independence. In vivo treatment with a CDK8/19 inhibitor Senexin B suppressed tumor growth and augmented the effects of fulvestrant in ER-positive breast cancer xenografts. These results identify CDK8 as a novel downstream mediator of ER and suggest the utility of CDK8 inhibitors for ER-positive breast cancer therapy.
Collapse
Affiliation(s)
- Martina S J McDermott
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Alexander A Chumanevich
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Chang-Uk Lim
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Jiaxin Liang
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Mengqian Chen
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Serena Altilia
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - David Oliver
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - James M Rae
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Michael Shtutman
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Hippokratis Kiaris
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, Semmelweis University 2nd Department of Pediatrics, Budapest, Hungary
| | - Igor B Roninson
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Eugenia V Broude
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, USA
| |
Collapse
|
180
|
Raj GV, Sareddy GR, Ma S, Lee TK, Viswanadhapalli S, Li R, Liu X, Murakami S, Chen CC, Lee WR, Mann M, Krishnan SR, Manandhar B, Gonugunta VK, Strand D, Tekmal RR, Ahn JM, Vadlamudi RK. Estrogen receptor coregulator binding modulators (ERXs) effectively target estrogen receptor positive human breast cancers. eLife 2017; 6. [PMID: 28786813 PMCID: PMC5548489 DOI: 10.7554/elife.26857] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 07/05/2017] [Indexed: 12/22/2022] Open
Abstract
The majority of human breast cancer is estrogen receptor alpha (ER) positive. While anti-estrogens/aromatase inhibitors are initially effective, resistance to these drugs commonly develops. Therapy-resistant tumors often retain ER signaling, via interaction with critical oncogenic coregulator proteins. To address these mechanisms of resistance, we have developed a novel ER coregulator binding modulator, ERX-11. ERX-11 interacts directly with ER and blocks the interaction between a subset of coregulators with both native and mutant forms of ER. ERX-11 effectively blocks ER-mediated oncogenic signaling and has potent anti-proliferative activity against therapy-sensitive and therapy-resistant human breast cancer cells. ERX-11 is orally bioavailable, with no overt signs of toxicity and potent activity in both murine xenograft and patient-derived breast tumor explant models. This first-in-class agent, with its novel mechanism of action of disrupting critical protein-protein interactions, overcomes the limitations of current therapies and may be clinically translatable for patients with therapy-sensitive and therapy-resistant breast cancers. DOI:http://dx.doi.org/10.7554/eLife.26857.001 Around 70% of breast cancers in women need one or both of the female hormones (estrogen and progesterone) to grow. To treat these 'hormone-dependent' cancers, patients receive drugs that either block the production of estrogen or directly target a receptor protein that senses estrogen in the cancer cells. Unfortunately, many breast cancers develop resistance to these drugs. This resistance is often caused by genetic mutations that alter the estrogen receptor; for example, the receptor may develop the ability to interact with other proteins in the cell known as coregulators to promote tumor growth. Developing new drugs that prevent estrogen receptors from interacting with coregulators may provide more options for treating hormone-dependent breast cancers. Here, Raj et al. developed a new small molecule named ERX-11 that is able to inhibit the growth of human breast cancer cells that are sensitive to existing drugs as well as cells that have become drug-resistant. For the experiments, hormone-dependent breast cancer cells from humans were transplanted into mice. This procedure usually causes the mice to develop tumors, but giving the mice ERX-11 by mouth stopped estrogen receptors from interacting with coregulators and blocked the growth of tumors. Furthermore, ERX-11 does not appear to have any toxic effects on the mice, indicating that it may also be safe for humans. The findings of Raj et al. suggest that ERX-11 is a promising new drug candidate for treating some breast cancers. The next steps are to examine the effects of ERX-11 on mice and other animals in more detail before deciding whether this molecule is suitable for clinical trials. In the longer term, molecules similar to ERX-11 could also be developed into drugs to treat other types of cancer that are also caused by abnormal interactions of coregulator proteins. DOI:http://dx.doi.org/10.7554/eLife.26857.002
Collapse
Affiliation(s)
- Ganesh V Raj
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Gangadhara Reddy Sareddy
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| | - Shihong Ma
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Tae-Kyung Lee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Suryavathi Viswanadhapalli
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Rui Li
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Xihui Liu
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Shino Murakami
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, United States.,Laboratory of Signaling and Gene Regulation, Cecil H and Ida Green Center for Reproductive Biology Sciences and Division of Basic Reproductive Biology Research, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chien-Cheng Chen
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Wan-Ru Lee
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Monica Mann
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Samaya Rajeshwari Krishnan
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Bikash Manandhar
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Vijay K Gonugunta
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States
| | - Douglas Strand
- Departments of Urology and Pharmacology, University of Texas Southwestern Medical Center at Dallas, Dallas, United States
| | - Rajeshwar Rao Tekmal
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| | - Jung-Mo Ahn
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, United States
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, San Antonio, United States.,CDP program, University of Texas Health Cancer Center, San Antonio, United States
| |
Collapse
|
181
|
Louka ML, Fawzy AM, Naiem AM, Elseknedy MF, Abdelhalim AE, Abdelghany MA. Vitamin D and K signaling pathways in hepatocellular carcinoma. Gene 2017; 629:108-116. [PMID: 28764978 DOI: 10.1016/j.gene.2017.07.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver malignancy, and is now the six most common in between malignancies. Early diagnosis of HCC with prompt treatment increases the opportunity of patients to survive. With the advances in understanding the molecular biology of HCC, new therapeutic strategies to treat HCC have emerged. There is a growing consensus that vitamins are important for the control of various cancers. Biochemical evidence clearly indicates that HCC cells are responsive to the inhibitory effect of vitamin D, vitamin D analogues and vitamin K. In this review, we summarize the mechanisms used by vitamin D and K to influence the development of HCC and the latest development of vitamin analogues for potential HCC therapy.
Collapse
Affiliation(s)
- Manal L Louka
- Medical Biochemistry Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Ahmed M Fawzy
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Abdelrahman M Naiem
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mustafa F Elseknedy
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Ahmed E Abdelhalim
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Mohamed A Abdelghany
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
| |
Collapse
|
182
|
Genome-wide identification of thyroid hormone receptor targets in the remodeling intestine during Xenopus tropicalis metamorphosis. Sci Rep 2017; 7:6414. [PMID: 28743885 PMCID: PMC5527017 DOI: 10.1038/s41598-017-06679-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/16/2017] [Indexed: 12/26/2022] Open
Abstract
Thyroid hormone (T3) affects development and metabolism in vertebrates. We have been studying intestinal remodeling during T3-dependent Xenopus metamorphosis as a model for organ maturation and formation of adult organ-specific stem cells during vertebrate postembryonic development, a period characterized by high levels of plasma T3. T3 is believed to affect development by regulating target gene transcription through T3 receptors (TRs). While many T3 response genes have been identified in different animal species, few have been shown to be direct target genes in vivo, especially during development. Here we generated a set of genomic microarray chips covering about 8000 bp flanking the predicted transcription start sites in Xenopus tropicalis for genome wide identification of TR binding sites. By using the intestine of premetamorphic tadpoles treated with or without T3 and for chromatin immunoprecipitation assays with these chips, we determined the genome-wide binding of TR in the control and T3-treated tadpole intestine. We further validated TR binding in vivo and analyzed the regulation of selected genes. We thus identified 278 candidate direct TR target genes. We further provided evidence that these genes are regulated by T3 and likely involved in the T3-induced formation of adult intestinal stem cells during metamorphosis.
Collapse
|
183
|
Yun SH, Park MG, Kim YM, Roh MS, Park JI. Expression of chicken ovalbumin upstream promoter-transcription factor II and liver X receptor as prognostic indicators for human colorectal cancer. Oncol Lett 2017; 14:4011-4020. [PMID: 28943908 PMCID: PMC5594251 DOI: 10.3892/ol.2017.6659] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/21/2017] [Indexed: 12/15/2022] Open
Abstract
Cholesterol increases the risk of colorectal cancer. Liver X receptor (LXR), retinoid X receptor (RXR)α and sterol regulatory element binding protein (SREBP)-1c are transcriptional regulators of lipid metabolism. Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) serves an essential role in angiogenesis and development, but its role in cancer is controversial. The expression of COUP-TFII, LXR, RXRα and SREBP-1c in colorectal cancer, as well as their association with clinicopathologic features, was assessed, and their utility as prognostic indicators in colorectal cancer evaluated. Colorectal cancer samples (n=707 patients) were analyzed for COUP-TII, LXR, RXRα and SREBP-1c expression by immunohistochemistry. Overall survival curves of patients with tumors expressing different levels of these proteins were produced and risk factors were assessed. Of the 707 patients, 32.7, 50.9, 56.4, and 41.7% were positive for COUP-TFII, LXR, RXRα, and SREBP-1c, respectively. The lack of COUP-TFII or LXR expression was associated with lower overall survival rates (P=0.0154 for COUP-TFII, and 0.0113 for LXR). Following adjustment for other clinical risk factors (age, sex, tumor size, grade, vascular invasion, and Tumor-Node-Metastasis stage), the lack of COUP-TFII or LXR expression was a negative independent prognostic factor for survival. The expression of COUP-TFII and LXR alone or in combination may be biomarkers to indicate a positive prognosis in patients with colorectal cancer.
Collapse
Affiliation(s)
- Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Min-Gyoung Park
- Department of Pathology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Yu-Mi Kim
- Department of Preventive Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Mee-Sook Roh
- Department of Pathology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| |
Collapse
|
184
|
Lou P, Li C, Shi L, Xia TS, Zhou W, Wu J, Zhou X, Li X, Wang Y, Wei JF, Ding Q. RNPC1 enhances progesterone receptor functions by regulating its mRNA stability in breast cancer. Oncotarget 2017; 8:16387-16400. [PMID: 27634883 PMCID: PMC5369970 DOI: 10.18632/oncotarget.12016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 07/26/2016] [Indexed: 02/04/2023] Open
Abstract
Progesterone receptor (PR) could activate transcriptional process involved in normal mammary gland proliferation and breast cancer development. Moreover, PR expression is an important marker of luminal breast cancer, which is associated with good prognosis and indicates better responding to endocrine therapies. The regulation of PR expression was studied mainly on its post-translational levels. In this study, we found PR was positively regulated by RNA-binding region-containing protein 1 (RNPC1), a RNA-binding protein, in PR positive breast cancer. Overexpression of RNPC1 increased, whereas knockdown of RNPC1 decreased, the level of PR protein and transcripts. Additionally, we demonstrated that RNPC1 could bind to PR mRNA via AU-rich elements (AREs) within PR 3′-untranslated region (3′-UTR) and then enhance PR mRNA stability. Moreover, we proved that progesterone-dependent PR functions which could induce breast cancer proliferation were enhanced by RNPC1, both in vitro and in vivo. Conclusively, we revealed a novel mechanism by which PR could be regulated by RNPC1 via stabilizing its mRNA.
Collapse
Affiliation(s)
- Peipei Lou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Chunlian Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Liang Shi
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Tian-Song Xia
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Wenbin Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Jing Wu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Xujie Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Xiaoxia Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Ying Wang
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu Province, 210000, China
| |
Collapse
|
185
|
Domonkos E, Borbélyová V, Csongová M, Bosý M, Kačmárová M, Ostatníková D, Hodosy J, Celec P. Sex differences and sex hormones in anxiety-like behavior of aging rats. Horm Behav 2017; 93:159-165. [PMID: 28576648 DOI: 10.1016/j.yhbeh.2017.05.019] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/17/2017] [Accepted: 05/28/2017] [Indexed: 01/21/2023]
Abstract
Sex differences in the prevalence of affective disorders might be attributable to different sex hormone milieu. The effects of short-term sex hormone deficiency on behavior, especially on anxiety have been studied in numerous animal experiments, mainly on young adult rats and mice. However, sex differences in aged animals and the effects of long-term hypogonadism are understudied. The aim of our study was to analyze sex differences in anxiety-like behavior in aged rats and to prove whether they can be attributed to endogenous sex hormone production in males. A battery of tests was performed to assess anxiety-like behavior in aged female, male and gonadectomized male rats castrated before puberty. In addition, the aged gonadectomized male rats were treated with a single injection of estradiol or testosterone or supplemented with estradiol for two-weeks. Female rats displayed a less anxious behavior than male rats in most of the conducted behavioral tests except the light-dark box. Long-term androgen deficiency decreased the sex difference in anxiety either partially (open field, PhenoTyper cage) or completely (elevated plus maze). Neither single injection of sex hormones, nor two-week supplementation of estradiol in gonadectomized aged male rats significantly affected their anxiety-like behavior in the elevated plus maze. In conclusion, our results confirm sex differences in anxiety in aged rats likely mediated by endogenous testosterone production in males. Whether long-term supplementation with exogenous sex hormones could affect anxiety-like behavior in elderly individuals remains to be elucidated.
Collapse
Affiliation(s)
- Emese Domonkos
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Veronika Borbélyová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Melinda Csongová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Martin Bosý
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Mária Kačmárová
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Daniela Ostatníková
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| | - Július Hodosy
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Institute of Pathophysiology, Faculty of Medicine, Comenius University, Bratislava, Slovakia; Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
186
|
Jeong J, Park S, An HT, Kang M, Ko J. Small leucine zipper protein functions as a negative regulator of estrogen receptor α in breast cancer. PLoS One 2017; 12:e0180197. [PMID: 28662179 PMCID: PMC5491147 DOI: 10.1371/journal.pone.0180197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/12/2017] [Indexed: 01/15/2023] Open
Abstract
The nuclear transcription factor estrogen receptor α (ERα) plays a critical role in breast cancer progression. ERα acts as an important growth stimulatory protein in breast cancer and the expression level of ERα is tightly related to the prognosis and treatment of patients. Small leucine zipper protein (sLZIP) functions as a transcriptional cofactor by binding to various nuclear receptors, including glucocorticoid receptor, androgen receptor, and peroxisome proliferator-activated receptor γ. However, the role of sLZIP in the regulation of ERα and its involvement in breast cancer progression is unknown. We found that sLZIP binds to ERα and represses the transcriptional activity of ERα in ERα-positive breast cancer cells. sLZIP also suppressed the expression of ERα target genes. sLZIP disrupted the binding of ERα to the estrogen response element of the target gene promoter, resulting in suppression of cell proliferation. sLZIP is a novel co-repressor of ERα, and plays a negative role in ERα-mediated cell proliferation in breast cancer.
Collapse
Affiliation(s)
- Juyeon Jeong
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Sodam Park
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Hyoung-Tae An
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Minsoo Kang
- Division of Life Sciences, Korea University, Seoul, South Korea
| | - Jesang Ko
- Division of Life Sciences, Korea University, Seoul, South Korea
- * E-mail:
| |
Collapse
|
187
|
Nongenomic Actions of 17-β Estradiol Restore Respiratory Neuroplasticity in Young Ovariectomized Female Rats. J Neurosci 2017; 37:6648-6660. [PMID: 28592693 DOI: 10.1523/jneurosci.0433-17.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 12/31/2022] Open
Abstract
Gonadal steroids modulate CNS plasticity, including phrenic long-term facilitation (pLTF), a form of spinal respiratory neuroplasticity resulting in increased phrenic nerve motor output following exposure to acute intermittent hypoxia (aIH; three 5 min episodes, 10.5% O2). Despite the importance of respiratory system neuroplasticity, and its dependence on estrogen in males, little is known about pLTF expression or mechanisms of estrogen signaling in females. Here, we tested the hypotheses that (1) pLTF expression in young, gonadally intact female rats would be expressed during estrous cycle stages in which 17β-estradiol (E2) is naturally high (e.g., proestrus vs estrus), (2) pLTF would be absent in ovariectomized (OVX) rats and in physiological conditions in which serum progesterone, but not E2, is elevated (e.g., lactating rats, 3-10 d postpartum), and (3) acute E2 administration would be sufficient to restore pLTF in OVX rats. Recordings of phrenic nerve activity in female Sprague Dawley rats (3-4 months) revealed a direct correlation between serum E2 levels and pLTF expression in cycling female rats. pLTF was abolished with OVX, but was re-established by acute E2 replacement (3 h, intraperitoneal). To identify underlying E2 signaling mechanisms, we intrathecally applied BSA-conjugated E2 over the spinal phrenic motor nucleus and found that pLTF expression was restored within 15 min, suggesting nongenomic E2 effects at membrane estrogen receptors. These data are the first to investigate the role of ovarian E2 in young cycling females, and to identify a role for nongenomic estrogen signaling in any form of respiratory system neuroplasticity.SIGNIFICANCE STATEMENT Exposure to acute intermittent hypoxia induces phrenic long-term facilitation (pLTF), a form of spinal respiratory motor plasticity that improves breathing in models of spinal cord injury. Although pathways leading to pLTF are well studied in males and estradiol (E2) is known to be required, it has seldom been investigated in females, and underlying mechanisms of E2 signaling are unknown in either sex. We found that while ovariectomy abolished pLTF, it could be restored by acute systemic E2, or by intraspinal application of the membrane-impermeable E2 (BSA-conjugated E2; 15 min). The ability of nongenomic estrogen signaling within the cervical spinal cord to recover respiratory neuroplasticity in disorders of respiratory insufficiency suggests that membrane estrogen receptors may represent novel therapeutic targets to restore breathing in both sexes.
Collapse
|
188
|
Oueslati M, Bittaieb I, Sassi N, Jemaa AB, Gamoudi A, Rahal K, Oueslati R. ERα and ERβ co-expression: An indicator of aggressive tumors and hormonal sensitivity. Oncol Lett 2017; 14:1675-1682. [PMID: 28808484 PMCID: PMC5542036 DOI: 10.3892/ol.2017.6314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/26/2017] [Indexed: 02/07/2023] Open
Abstract
The estrogen receptors (ERs) ERα and ERβ are important factors in breast cancer progression. Nevertheless, the molecular interplay between ERα and ERβ and its clinical significance in breast cancer is controversial. The establishment of a clear association is required; therefore, the current study analyzed the expression patterns of ERα and ERβ in 32 breast tumor tissues using reverse transcription-quantitative polymerase chain reaction. Furthermore, human epidermal growth factor receptor 2 (HER2) and the Ki-67 status were detected by immunohistochemistry. The results revealed that the ERα and ERβ expression rates recorded were 68 and 65%, respectively. The ERα:ERβ ratio exhibited a decline along with disease progression. ERα and ERβ were found to be negatively correlated with HER2 status but positively correlated with Ki-67. Co-expression of ERα and ERβ was associated with breast cancer aggressiveness, including higher histological grade and positive nodal status, which commonly occur following the menopause. In addition, in cases where ERβ was coexpressed with ERα, HER2 expression was frequently found to be negative, whereas the Ki-67 index was upregulated. These data suggest that ERα and ERβ co-expression may be an indicator of tumor aggressiveness and the sensitivity of hormonal therapy via the downregulation of HER2.
Collapse
Affiliation(s)
- Mohamed Oueslati
- Unit of Immunology, Environmental Microbiology and Carcinogenesis, Faculty of Sciences of Bizerte, University of Carthage, Zarzouna 7021, Tunisia
| | - Ilhem Bittaieb
- Pathological Anatomy Service, Saleh Azeiz Oncology Institute, Beb Saadoun, Tunis 1006, Tunisia
| | - Nadia Sassi
- Immuno-Rheumatology Research laboratory, Rheumatology Department, La Rabta Hospital, Tunis 1007, Tunisia
| | - Awatef Ben Jemaa
- Unit of Immunology, Environmental Microbiology and Carcinogenesis, Faculty of Sciences of Bizerte, University of Carthage, Zarzouna 7021, Tunisia
| | - Amor Gamoudi
- Pathological Anatomy Service, Saleh Azeiz Oncology Institute, Beb Saadoun, Tunis 1006, Tunisia
| | - Khaled Rahal
- Cancer Surgery Service, Saleh Azeiz Oncology Institute, Beb Saadoun, Tunis 1006, Tunisia
| | - Ridha Oueslati
- Unit of Immunology, Environmental Microbiology and Carcinogenesis, Faculty of Sciences of Bizerte, University of Carthage, Zarzouna 7021, Tunisia
| |
Collapse
|
189
|
Campos-Parra AD, Mitznahuatl GC, Pedroza-Torres A, Romo RV, Reyes FIP, López-Urrutia E, Pérez-Plasencia C. Micro-RNAs as Potential Predictors of Response to Breast Cancer Systemic Therapy: Future Clinical Implications. Int J Mol Sci 2017; 18:E1182. [PMID: 28574440 PMCID: PMC5486005 DOI: 10.3390/ijms18061182] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/24/2017] [Accepted: 05/27/2017] [Indexed: 12/11/2022] Open
Abstract
Despite advances in diagnosis and new treatments such as targeted therapies, breast cancer (BC) is still the most prevalent tumor in women worldwide and the leading cause of death. The principal obstacle for successful BC treatment is the acquired or de novo resistance of the tumors to the systemic therapy (chemotherapy, endocrine, and targeted therapies) that patients receive. In the era of personalized treatment, several studies have focused on the search for biomarkers capable of predicting the response to this therapy; microRNAs (miRNAs) stand out among these markers due to their broad spectrum or potential clinical applications. miRNAs are conserved small non-coding RNAs that act as negative regulators of gene expression playing an important role in several cellular processes, such as cell proliferation, autophagy, genomic stability, and apoptosis. We reviewed recent data that describe the role of miRNAs as potential predictors of response to systemic treatments in BC. Furthermore, upon analyzing the collected published information, we noticed that the overexpression of miR-155, miR-222, miR-125b, and miR-21 predicts the resistance to the most common systemic treatments; nonetheless, the function of these particular miRNAs must be carefully studied and further analyses are still necessary to increase knowledge about their role and future potential clinical uses in BC.
Collapse
Affiliation(s)
- Alma D Campos-Parra
- Laboratorio de Genomica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, C.P. 14080 Tlalpan, Ciudad de México, Mexico.
| | - Gerardo Cuamani Mitznahuatl
- Laboratorio de Genomica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, C.P. 14080 Tlalpan, Ciudad de México, Mexico.
| | - Abraham Pedroza-Torres
- Laboratorio de Genomica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, C.P. 14080 Tlalpan, Ciudad de México, Mexico.
- CATEDRA-CONACyT, Av. De los Insurgente Sur 1582, Col. Crédito Constructor., C.P. 03940 Benito Juárez, Ciudad de México, Mexico.
| | - Rafael Vázquez Romo
- Departamento de Cirugia de Tumores mamarios, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, C.P. 14080 Tlalpan, Ciudad de México, Mexico.
| | - Fany Iris Porras Reyes
- Servicio de Anatomia Patologica, Instituto Nacional de Cancerología (INCan), Av. San Fernando 22, Col. Sección XVI, C.P. 14080 Tlalpan, Ciudad de México, Mexico.
| | - Eduardo López-Urrutia
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de Mexico (UNAM), Av. De Los Barrios 1, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, C.P. 54090 Tlalnepantla, México, Mexico.
| | - Carlos Pérez-Plasencia
- Unidad de Biomedicina, FES-IZTACALA, Universidad Nacional Autónoma de Mexico (UNAM), Av. De Los Barrios 1, Los Reyes Ixtacala, Hab Los Reyes Ixtacala Barrio de los Árboles/Barrio de los Héroes, C.P. 54090 Tlalnepantla, México, Mexico.
| |
Collapse
|
190
|
Simonini S, Deb J, Moubayidin L, Stephenson P, Valluru M, Freire-Rios A, Sorefan K, Weijers D, Friml J, Østergaard L. A noncanonical auxin-sensing mechanism is required for organ morphogenesis in Arabidopsis. Genes Dev 2017; 30:2286-2296. [PMID: 27898393 PMCID: PMC5110995 DOI: 10.1101/gad.285361.116] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/13/2016] [Indexed: 01/18/2023]
Abstract
Tissue patterning in multicellular organisms is the output of precise spatio-temporal regulation of gene expression coupled with changes in hormone dynamics. In plants, the hormone auxin regulates growth and development at every stage of a plant's life cycle. Auxin signaling occurs through binding of the auxin molecule to a TIR1/AFB F-box ubiquitin ligase, allowing interaction with Aux/IAA transcriptional repressor proteins. These are subsequently ubiquitinated and degraded via the 26S proteasome, leading to derepression of auxin response factors (ARFs). How auxin is able to elicit such a diverse range of developmental responses through a single signaling module has not yet been resolved. Here we present an alternative auxin-sensing mechanism in which the ARF ARF3/ETTIN controls gene expression through interactions with process-specific transcription factors. This noncanonical hormone-sensing mechanism exhibits strong preference for the naturally occurring auxin indole 3-acetic acid (IAA) and is important for coordinating growth and patterning in diverse developmental contexts such as gynoecium morphogenesis, lateral root emergence, ovule development, and primary branch formation. Disrupting this IAA-sensing ability induces morphological aberrations with consequences for plant fitness. Therefore, our findings introduce a novel transcription factor-based mechanism of hormone perception in plants.
Collapse
Affiliation(s)
- Sara Simonini
- Department of Crop Genetics, John Innes Centre, Norwich NR4 7UH, United Kingdom
| | - Joyita Deb
- Department of Crop Genetics, John Innes Centre, Norwich NR4 7UH, United Kingdom
| | - Laila Moubayidin
- Department of Crop Genetics, John Innes Centre, Norwich NR4 7UH, United Kingdom
| | - Pauline Stephenson
- Department of Crop Genetics, John Innes Centre, Norwich NR4 7UH, United Kingdom
| | - Manoj Valluru
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Alejandra Freire-Rios
- Laboratory of Biochemistry, Wageningen University, 6703 HA Wageningen, the Netherlands
| | - Karim Sorefan
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield S10 2TN, United Kingdom
| | - Dolf Weijers
- Laboratory of Biochemistry, Wageningen University, 6703 HA Wageningen, the Netherlands
| | - Jiří Friml
- Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Lars Østergaard
- Department of Crop Genetics, John Innes Centre, Norwich NR4 7UH, United Kingdom
| |
Collapse
|
191
|
Fernandez EJ, Gahlot V, Rodriguez C, Amburn J. DNA-induced unfolding of the thyroid hormone receptor α A/B domain through allostery. FEBS Open Bio 2017; 7:854-864. [PMID: 28593140 PMCID: PMC5458466 DOI: 10.1002/2211-5463.12229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 01/22/2023] Open
Affiliation(s)
- Elias J. Fernandez
- Biochemistry & Cellular and Molecular Biology; University of Tennessee; Knoxville TN USA
| | - Vandna Gahlot
- Biochemistry & Cellular and Molecular Biology; University of Tennessee; Knoxville TN USA
| | - Celeste Rodriguez
- Biochemistry & Cellular and Molecular Biology; University of Tennessee; Knoxville TN USA
| | - Jacob Amburn
- Biochemistry & Cellular and Molecular Biology; University of Tennessee; Knoxville TN USA
| |
Collapse
|
192
|
Donoghue LJ, Neufeld TI, Li Y, Arao Y, Coons LA, Korach KS. Differential Activation of a Mouse Estrogen Receptor β Isoform (mERβ2) with Endocrine-Disrupting Chemicals (EDCs). ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:634-642. [PMID: 27634370 PMCID: PMC5381991 DOI: 10.1289/ehp396] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/03/2016] [Accepted: 08/17/2016] [Indexed: 05/09/2023]
Abstract
BACKGROUND Endocrine-disrupting chemicals (EDCs) are suspected of altering estrogenic signaling through estrogen receptor (ER) α or β (mERβ1 in mice). Several EDC effects have been reported in animal studies and extrapolated to human studies. Unlike humans, rodents express a novel isoform of ERβ (mERβ2) with a modified ligand-binding domain sequence. EDC activity through this isoform remains uncharacterized. OBJECTIVES We identified the expression pattern of mERβ2 in mouse tissues and assessed the estrogenic activity of EDCs through mERβ2. METHODS mERβ2 mRNA expression was measured in mouse tissues. HepG2 cells were used to assess the transactivation activity of mERβ isoforms with EDCs and ER co-activators. 293A cells transiently transfected with mER isoforms were used to detect EDC-mediated changes in endogenous ER target gene expression. RESULTS Expression of mERβ2 mRNA was detected in mouse reproductive tissues (ovary, testis, and prostate) and lung and colon tissues from both female and male mice. Five (E2, DES, DPN, BPAF, Coum, 1-BP) of 16 compounds tested by reporter assay had estrogenic activity through mERβ2. mERβ2 had a compound-specific negative effect on ERβ/ligand-mediated activity and ER target genes when co-expressed with mERβ1. mERβ2 recruited coactivators SRC2 or SRC3 in the presence of EDCs, but showed less recruitment than mERβ1. CONCLUSION mERβ2 showed weaker estrogenic activity than mERβ1 in our in vitro system, and can dampen mERβ1 activity. In vivo models of EDC activity and ER-mediated toxicity should consider the role of mERβ2, as rodent tissue responses involving mERβ2 may not be reproduced in human biology.
Collapse
Affiliation(s)
| | | | | | | | | | - Kenneth S. Korach
- Address correspondence to K.S. Korach, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., P.O. Box 12233, Research Triangle Park, NC 27709 USA. Telephone: (919) 541-3512. E-mail:
| |
Collapse
|
193
|
Hepatic effects of tartrazine (E 102) after systemic exposure are independent of oestrogen receptor interactions in the mouse. Toxicol Lett 2017; 273:55-68. [PMID: 28356238 PMCID: PMC5429395 DOI: 10.1016/j.toxlet.2017.03.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/23/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022]
Abstract
Systemic exposure to tartrazine results in hepatic periportal recruitment of inflammatory cells, increased serum alkaline phosphatase activity and mild hepatic periportal fibrosis. Tartrazine, its sulphonated metabolites and a common contaminant of the food additive do not interact with murine oestrogen receptors. Systemic exposure does not have an oestrogenic effect in mouse in vivo. Tartrazine, its sulphonated metabolites and a common contaminant of the food additive inhibited sulphotransferase, which may account for its hepatic effects after systemic exposure. The hepatic effects of tartrazine do not occur in mice – with or without co-administration of alcohol – after oral exposure to tartrazine.
Tartrazine is a food colour that activates the transcriptional function of the human oestrogen receptor alpha in an in vitro cell model. Since oestrogens are cholestatic, we hypothesised tartrazine will cause periportal injury to the liver in vivo. To test this hypothesis, tartrazine was initially administered systemically to mice resulting in a periportal recruitment of inflammatory cells, increased serum alkaline phosphatase activity and mild periportal fibrosis. To determine whether an oestrogenic effect may be a key event in this response, tartrazine, sulphonated metabolites and a food additive contaminant were screened for their ability to interact with murine oestrogen receptors. In all cases, there were no interactions as agonists or antagonists and further, no oestrogenicity was observed with tartrazine in an in vivo uterine growth assay. To examine the relevance of the hepatic effects of tartrazine to its use as a food additive, tartrazine was orally administered to transgenic NF-κB-Luc mice. Pre- and concurrent oral treatment with alcohol was incorporated given its potential to promote gut permeability and hepatic inflammation. Tartrazine alone induced NF- κB activities in the colon and liver but there was no periportal recruitment of inflammatory cells or fibrosis. Tartrazine, its sulphonated metabolites and the contaminant inhibited sulphotransferase activities in murine hepatic S9 extracts. Given the role of sulfotransferases in bile acid excretion, the initiating event giving rise to periportal inflammation and subsequent hepatic pathology through systemic tartrazine exposure is therefore potentially associated an inhibition of bile acid sulphation and excretion and not on oestrogen receptor-mediated transcriptional function. However, these effects were restricted to systemic exposures to tartrazine and did not occur to any significant effect after oral exposure.
Collapse
|
194
|
Regitz-Zagrosek V, Kararigas G. Mechanistic Pathways of Sex Differences in Cardiovascular Disease. Physiol Rev 2017; 97:1-37. [PMID: 27807199 DOI: 10.1152/physrev.00021.2015] [Citation(s) in RCA: 458] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Major differences between men and women exist in epidemiology, manifestation, pathophysiology, treatment, and outcome of cardiovascular diseases (CVD), such as coronary artery disease, pressure overload, hypertension, cardiomyopathy, and heart failure. Corresponding sex differences have been studied in a number of animal models, and mechanistic investigations have been undertaken to analyze the observed sex differences. We summarize the biological mechanisms of sex differences in CVD focusing on three main areas, i.e., genetic mechanisms, epigenetic mechanisms, as well as sex hormones and their receptors. We discuss relevant subtypes of sex hormone receptors, as well as genomic and nongenomic, activational and organizational effects of sex hormones. We describe the interaction of sex hormones with intracellular signaling relevant for cardiovascular cells and the cardiovascular system. Sex, sex hormones, and their receptors may affect a number of cellular processes by their synergistic action on multiple targets. We discuss in detail sex differences in organelle function and in biological processes. We conclude that there is a need for a more detailed understanding of sex differences and their underlying mechanisms, which holds the potential to design new drugs that target sex-specific cardiovascular mechanisms and affect phenotypes. The comparison of both sexes may lead to the identification of protective or maladaptive mechanisms in one sex that could serve as a novel therapeutic target in one sex or in both.
Collapse
Affiliation(s)
- Vera Regitz-Zagrosek
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Georgios Kararigas
- Institute of Gender in Medicine & Center for Cardiovascular Research, Charite University Hospital, and DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
195
|
Pluchino N, Russo M, Genazzani AR. The fetal brain: role of progesterone and allopregnanolone. Horm Mol Biol Clin Investig 2017; 27:29-34X. [PMID: 27442421 DOI: 10.1515/hmbci-2016-0020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/29/2016] [Indexed: 01/14/2023]
Abstract
Progesterone and allopregnanolone have crucial and different roles in brain development, function and recovery after injury. Pregnancy is characterized by an increased synthesis of progesterone and its neuro-active metabolites by the placenta, maternal and fetal brain. This supports the critical role of these steroids in maternal brain adaptation during pregnancy and development of the fetal brain. Moreover, allopregnanolone may play a brain-protective role during complications of pregnancy, complications of pregnancy, such as preterm delivery or intrauterine growth restriction (IUGR), by reducing the impact of hypoxia and excitotoxic brain damage or impairment myelination. Behavioral consequences of altered progesterone/allopregnanolone fetal brain programming have also been hypothesized, although further evidence is needed. New potential applications of allopregnanolone as a treatment strategy have also been proposed, addressing unmet clinical needs in perinatal care.
Collapse
|
196
|
Chiang KC, Yeh TS, Huang CC, Chang YC, Juang HH, Cheng CT, Pang JHS, Hsu JT, Takano M, Chen TC, Kittaka A, Hsiao M, Yeh CN. MART-10 represses cholangiocarcinoma cell growth and high vitamin D receptor expression indicates better prognosis for cholangiocarcinoma. Sci Rep 2017; 7:43773. [PMID: 28256614 PMCID: PMC5335655 DOI: 10.1038/srep43773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/30/2017] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a devastating disease due to no effective treatments available. Since the non-mineral functions of vitamin D emerges, 1α,25(OH)2D3, the active form of vitamin D, has been applied in anti-cancer researches. In this study, we demonstrated that both the 1α,25(OH)2D3 analog, MART-10, and 1α,25(OH)2D3 possessed anti-growth effect on human CCA cells with MART-10 much more potent than 1α,25(OH)2D3. The growth inhibition of both drugs were mediated by induction of G0/G1 cell cycle arrest through upregulation of p27 and downregulation of CDK4, CDK6, and cyclin D3. Human neutrophil gelatinase associated lipocalin (NGAL) was found to be involved in 1α,25(OH)2D3 and MART-10 meditated growth inhibition for CCA as knockdown of NGAL decreased Ki-67 expression in SNU308 cells and rendered SNU308 cells less responsive to 1α,25(OH)2D3 and MART-10 treatment. Vitamin D receptor (VDR) knockdown partly abolished MART-10-induced inhibition of NGAL and cell growth in SNU308 cells. The xenograft animal study demonstrated MART-10 could effectively repressed CCA growth in vivo without inducing obvious side effects. The IHC examination of human CCA specimen for VDR revealed that higher VDR expression was linked with better prognosis. Collectively, our results suggest that MART-10 could be a promising regimen for CCA treatment.
Collapse
Affiliation(s)
- Kun-Chun Chiang
- General Surgery Department, Chang Gung Memorial Hospital, Chang Gung University, Keelung, R.O.C, Taiwan
- Director of Zebrafish center of Keelung Chang Gung Memorial Hospital, R.O.C, Taiwan
| | - Ta-Sen Yeh
- General Surgery Department and Liver research center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-Shan, Taoyuan, R.O.C, Taiwan
| | - Cheng-Cheng Huang
- Department of Pathology, Chang Gung Memorial Hospital, 222, Mai-Chin Road, Keelung, R.O.C, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, R.O.C, Taiwan
| | - Horng-Heng Juang
- Department of Anatomy, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan, 333, R.O.C, Taiwan
| | - Chi-Tung Cheng
- General Surgery Department and Liver research center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-Shan, Taoyuan, R.O.C, Taiwan
| | - Jong-Hwei S. Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan, R.O.C, Taiwan
| | - Jun-Te Hsu
- General Surgery Department and Liver research center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-Shan, Taoyuan, R.O.C, Taiwan
| | - Masashi Takano
- Faculty of Pharmaceutical Sciences, Teikyo University, Sagamihara, Kanagawa, 252-5195, Japan
| | - Tai C. Chen
- Boston University School of Medicine, M-1022, 715 Albany Street, Boston, MA 02118, USA
| | - Atsushi Kittaka
- Faculty of Pharmaceutical Sciences, Teikyo University, Sagamihara, Kanagawa, 252-5195, Japan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, R.O.C, Taiwan
| | - Chun-Nan Yeh
- General Surgery Department and Liver research center, Chang Gung Memorial Hospital, Chang Gung University, Kwei-Shan, Taoyuan, R.O.C, Taiwan
| |
Collapse
|
197
|
O'Shea LC, Daly E, Hensey C, Fair T. ATRX is a novel progesterone-regulated protein and biomarker of low developmental potential in mammalian oocytes. Reproduction 2017; 153:671-682. [PMID: 28250240 DOI: 10.1530/rep-16-0443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 02/03/2017] [Accepted: 02/28/2017] [Indexed: 01/09/2023]
Abstract
A multi-species meta-analysis of published transcriptomic data from models of oocyte competence identified the chromatin remodelling factor ATRX as a putative biomarker of oocyte competence. The objective of the current study was to test the hypothesis that ATRX protein expression by cumulus-oocyte complexes (COCs) reflects their intrinsic quality and developmental potential. In excess of 10,000 bovine COCs were utilised to test our hypothesis. COCs were in vitro matured (IVM) under conditions associated with reduced developmental potential: IVM in the presence or absence of (1) progesterone synthesis inhibitor (Trilostane); (2) nuclear progesterone receptor inhibitor (Aglepristone) or (3) an inducer of DNA damage (Staurosporine). ATRX protein expression and localisation were determined using immunocytochemistry and Western blot analysis. A proportion of COCs matured in the presence or absence of Trilostane was in vitro fertilised and cultured, and subsequent embryo development characteristics were analysed. In addition, ATRX expression was investigated in 40 human germinal vesicle-stage COCs. Our results showed that ATRX is expressed in human and bovine germinal vesicle oocytes and cumulus cells. In bovine, expression decreases after IVM. However, this decline is not observed in COCs matured under sub-optimal conditions. Blastocyst development rate and cell number are decreased, whereas the incidence of abnormal metaphase phase spindle and chromosome alignment are increased, after IVM in the presence of Trilostane (P < 0.05). In conclusion, localisation of ATRX to the cumulus cell nuclei and oocyte chromatin, after IVM, is associated with poor oocyte quality and low developmental potential. Furthermore, ATRX is dynamically regulated in response to progesterone signalling.
Collapse
Affiliation(s)
- Lynne C O'Shea
- School of Agriculture and Food Sciences .,School of Medicine
| | | | - Carmel Hensey
- School of Bimolecular and Biomedical ScienceUniversity College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
198
|
Obinata D, Takayama K, Takahashi S, Inoue S. Crosstalk of the Androgen Receptor with Transcriptional Collaborators: Potential Therapeutic Targets for Castration-Resistant Prostate Cancer. Cancers (Basel) 2017; 9:E22. [PMID: 28264478 PMCID: PMC5366817 DOI: 10.3390/cancers9030022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is the second leading cause of death from cancer among males in Western countries. It is also the most commonly diagnosed male cancer in Japan. The progression of prostate cancer is mainly influenced by androgens and the androgen receptor (AR). Androgen deprivation therapy is an established therapy for advanced prostate cancer; however, prostate cancers frequently develop resistance to low testosterone levels and progress to the fatal stage called castration-resistant prostate cancer (CRPC). Surprisingly, AR and the AR signaling pathway are still activated in most CRPC cases. To overcome this problem, abiraterone acetate and enzalutamide were introduced for the treatment of CRPC. Despite the impact of these drugs on prolonged survival, CRPC acquires further resistance to keep the AR pathway activated. Functional molecular studies have shown that some of the AR collaborative transcription factors (TFs), including octamer transcription factor (OCT1), GATA binding protein 2 (GATA2) and forkhead box A1 (FOXA1), still stimulate AR activity in the castration-resistant state. Therefore, elucidating the crosstalk between the AR and collaborative TFs on the AR pathway is critical for developing new strategies for the treatment of CRPC. Recently, many compounds targeting this pathway have been developed for treating CRPC. In this review, we summarize the AR signaling pathway in terms of AR collaborators and focus on pyrrole-imidazole (PI) polyamide as a candidate compound for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Daisuke Obinata
- Department of Urology, Nihon University School of Medicine, Tokyo 173-8610, Japan.
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| | - Kenichi Takayama
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Satoshi Inoue
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Saitama 350-1241, Japan.
| |
Collapse
|
199
|
Ponce-Ruiz N, Murillo-González FE, Rojas-García AE, Mackness M, Bernal-Hernández YY, Barrón-Vivanco BS, González-Arias CA, Medina-Díaz IM. Transcriptional regulation of human Paraoxonase 1 by nuclear receptors. Chem Biol Interact 2017; 268:77-84. [PMID: 28223025 DOI: 10.1016/j.cbi.2017.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 02/10/2017] [Indexed: 01/08/2023]
Abstract
Paraoxonase 1 (PON1) is a calcium-dependent lactonase synthesized primarily in the liver and secreted into the plasma, where it is associates with high density lipoproteins (HDL). PON1 acts as antioxidant preventing low-density lipoprotein (LDL) oxidation, a process considered critical in the initiation and progression of atherosclerosis. Additionally, PON1 hydrolyzes and detoxifies some toxic metabolites of organophosphorus compounds (OPs). Thus, PON1 activity and expression levels are important for determining susceptibility to OPs intoxication and risk of developing diseases related to inflammation and oxidative stress. Increasing evidence has demonstrated the modulation of PON1 expression by many factors is due to interaction with nuclear receptors (NRs). Here, we briefly review the studies in this area and discuss the role of nuclear receptors in the regulation of PON1 expression, as well as how understanding these mechanisms may allow us to manipulate PON1 levels to improve drug efficacy and treat disease.
Collapse
Affiliation(s)
- N Ponce-Ruiz
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico; Posgrado en Ciencias Biológico Agropecuarias, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico.
| | - F E Murillo-González
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico; Posgrado en Ciencias Biológico Agropecuarias, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico.
| | - A E Rojas-García
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico.
| | - Mike Mackness
- Institute of Cardiovascular Sciences, Manchester, United Kingdom.
| | - Y Y Bernal-Hernández
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico.
| | - B S Barrón-Vivanco
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico.
| | - C A González-Arias
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico.
| | - I M Medina-Díaz
- Universidad Autónoma de Nayarit, Laboratorio de Contaminación y Toxicología, Secretaría de Investigación y Posgrado, Nayarit, Mexico.
| |
Collapse
|
200
|
Male reproductive toxicity of CrVI: In-utero exposure to CrVI at the critical window of testis differentiation represses the expression of Sertoli cell tight junction proteins and hormone receptors in adult F 1 progeny rats. Reprod Toxicol 2017; 69:84-98. [PMID: 28192182 DOI: 10.1016/j.reprotox.2017.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 01/19/2017] [Accepted: 02/08/2017] [Indexed: 11/24/2022]
Abstract
The effect of gestational exposure to CrVI (occupational/environmental pollutant and target to Sertoli cells(SC)) was tested in a rat model during the testicular differentiation from the bipotential gonad may interrupt spermatogenesis by disrupting SC tight junctions(TJ) and it's proteins and hormone receptors. Pregnant Wistar rats were exposed to 50/100/200ppm CrVI through drinking water during embryonic days 9-14. On Postnatal day 120, testes were subjected to ion exchange chromatographic analysis and revealed increased level of CrIII in SCs and germ cells, serum and testicular interstitial fluid(TIF). Microscopic analyses showed seminiferous tubules atrophy and disruption of SC TJ, which also recorded decreased testosterone in TIF. mRNA and Protein expression analyses attested decreased level of Fshr, Ar, occludin and claudin-11 in SCs. Immunofluorescent detection revealed weak signal of TJ proteins. Taken together, we concluded that gestational exposure to CrVI interferes with the expression of SC TJ proteins due to attenuated expression of hormone receptors.
Collapse
|