11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      A comprehensive data mining study shows that most nuclear receptors act as newly proposed homeostasis-associated molecular pattern receptors

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Nuclear receptors (NRs) can regulate gene expression; therefore, they are classified as transcription factors. Despite the extensive research carried out on NRs, still several issues including (1) the expression profile of NRs in human tissues, (2) how the NR expression is modulated during atherosclerosis and metabolic diseases, and (3) the overview of the role of NRs in inflammatory conditions are not fully understood.

          Methods

          To determine whether and how the expression of NRs are regulated in physiological/pathological conditions, we took an experimental database analysis to determine expression of all 48 known NRs in 21 human and 17 murine tissues as well as in pathological conditions.

          Results

          We made the following significant findings: (1) NRs are differentially expressed in tissues, which may be under regulation by oxygen sensors, angiogenesis pathway, stem cell master regulators, inflammasomes, and tissue hypo-/hypermethylation indexes; (2) NR sequence mutations are associated with increased risks for development of cancers and metabolic, cardiovascular, and autoimmune diseases; (3) NRs have less tendency to be upregulated than downregulated in cancers, and autoimmune and metabolic diseases, which may be regulated by inflammation pathways and mitochondrial energy enzymes; and (4) the innate immune sensor inflammasome/caspase-1 pathway regulates the expression of most NRs.

          Conclusions

          Based on our findings, we propose a new paradigm that most nuclear receptors are anti-inflammatory homeostasis-associated molecular pattern receptors (HAMPRs). Our results have provided a novel insight on NRs as therapeutic targets in metabolic diseases, inflammations, and malignancies.

          Electronic supplementary material

          The online version of this article (10.1186/s13045-017-0526-8) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references81

          • Record: found
          • Abstract: found
          • Article: not found

          The steroid and thyroid hormone receptor superfamily.

          Analyses of steroid receptors are important for understanding molecular details of transcriptional control, as well as providing insight as to how an individual transacting factor contributes to cell identity and function. These studies have led to the identification of a superfamily of regulatory proteins that include receptors for thyroid hormone and the vertebrate morphogen retinoic acid. Although animals employ complex and often distinct ways to control their physiology and development, the discovery of receptor-related molecules in a wide range of species suggests that mechanisms underlying morphogenesis and homeostasis may be more ubiquitous than previously expected.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta.

            The rat estrogen receptor (ER) exists as two subtypes, ER alpha and ER beta, which differ in the C-terminal ligand binding domain and in the N-terminal transactivation domain. In this study we investigated the messenger RNA expression of both ER subtypes in rat tissues by RT-PCR and compared the ligand binding specificity of the ER subtypes. Saturation ligand binding analysis of in vitro synthesized human ER alpha and rat ER beta protein revealed a single binding component for 16 alpha-iodo-17 beta-estradiol with high affinity [dissociation constant (Kd) = 0.1 nM for ER alpha protein and 0.4 nM for ER beta protein]. Most estrogenic substances or estrogenic antagonists compete with 16 alpha-[125I]iodo-17 beta-estradiol for binding to both ER subtypes in a very similar preference and degree; that is, diethylstilbestrol > hexestrol > dienestrol > 4-OH-tamoxifen > 17 beta-estradiol > coumestrol, ICI-164384 > estrone, 17 alpha-estradiol > nafoxidine, moxestrol > clomifene > estriol, 4-OH-estradiol > tamoxifen, 2-OH-estradiol, 5-androstene-3 beta, 17 beta-diol, genistein for the ER alpha protein and dienestrol > 4-OH-tamoxifen > diethylstilbestrol > hexestrol > coumestrol, ICI-164384 > 17 beta-estradiol > estrone, genistein > estriol > nafoxidine, 5-androstene-3 beta, 17 beta-diol > 17 alpha-estradiol, clomifene, 2-OH-estradiol > 4-OH-estradiol, tamoxifen, moxestrol for the ER beta protein. The rat tissue distribution and/or the relative level of ER alpha and ER beta expression seems to be quite different, i.e. moderate to high expression in uterus, testis, pituitary, ovary, kidney, epididymis, and adrenal for ER alpha and prostate, ovary, lung, bladder, brain, uterus, and testis for ER beta. The described differences between the ER subtypes in relative ligand binding affinity and tissue distribution could contribute to the selective action of ER agonists and antagonists in different tissues.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network.

              In multicellular organisms, the ability to regulate reproduction, development, and nutrient utilization coincided with the evolution of nuclear receptors (NRs), transcription factors that utilize lipophilic ligands to mediate their function. Studying the expression profile of NRs offers a simple, powerful way to obtain highly relational information about their physiologic functions as individual proteins and as a superfamily. We surveyed the expression of all 49 mouse NR mRNAs in 39 tissues, representing diverse anatomical systems. The resulting data set uncovers several NR clades whose patterns of expression indicate their ability to coordinate the transcriptional programs necessary to affect distinct physiologic pathways. Remarkably, this regulatory network divides along the following two physiologic paradigms: (1) reproduction, development, and growth and (2) nutrient uptake, metabolism, and excretion. These data reveal a hierarchical transcriptional circuitry that extends beyond individual tissues to form a meganetwork governing physiology on an organismal scale.
                Bookmark

                Author and article information

                Contributors
                wlq8360@163.com
                gayani@temple.edu
                1091251425@qq.com
                tanhm@mail.sysu.edu.cn
                tug65470@temple.edu
                tug36537@temple.edu
                tuf43098@temple.edu
                tuf07694@temple.edu
                tue54600@temple.edu
                tue44606@temple.edu
                Teodoro.Bottiglieri@BSWHealth.org
                tuf22569@temple.edu
                tuf65151@temple.edu
                yingyang1612@gmail.com
                gayanikn@gmail.com
                745950710@qq.com
                hangxi@temple.edu
                43362230@qq.com
                junyu@temple.edu
                Eric.Choi@tuhs.temple.edu
                xiaoshumenfan@126.com
                hongw@temple.edu
                xfyang@temple.edu
                Journal
                J Hematol Oncol
                J Hematol Oncol
                Journal of Hematology & Oncology
                BioMed Central (London )
                1756-8722
                24 October 2017
                24 October 2017
                2017
                : 10
                : 168
                Affiliations
                [1 ]GRID grid.412455.3, Department of Cardiovascular Medicine, , The Second Affiliated Hospital of Nanchang University, ; Nanchang, Jiangxi 330006 China
                [2 ]ISNI 0000 0001 2248 3398, GRID grid.264727.2, Center for Metabolic Disease Research, , Lewis Katz School of Medicine at Temple University, ; Philadelphia, PA 19140 USA
                [3 ]ISNI 0000 0001 2248 3398, GRID grid.264727.2, Centers for Cardiovascular Research and Thrombosis Research, , Lewis Katz School of Medicine at Temple University, ; Philadelphia, PA 19140 USA
                [4 ]ISNI 0000 0001 2248 3398, GRID grid.264727.2, Department of Pharmacology, , Lewis Katz School of Medicine at Temple University, ; Philadelphia, PA 19140 USA
                [5 ]ISNI 0000 0001 2248 3398, GRID grid.264727.2, Department of Surgery, , Lewis Katz School of Medicine at Temple University, ; Philadelphia, PA 19140 USA
                [6 ]GRID grid.414902.a, Department of Cardiovascular Medicine, , The First Affiliated Hospital of Kunming Medical University, ; Kunming, Yunnan 650032 China
                [7 ]ISNI 0000 0004 1799 374X, GRID grid.417295.c, Department of Ultrasound, , Xijing Hospital and Fourth Military Medical University, ; Xi’an, Shaanxi 710032 China
                [8 ]ISNI 0000 0001 2360 039X, GRID grid.12981.33, Department of Pathophysiology, Zhongshan School of Medicine, , Sun Yat-sen University, ; Guangzhou, Guangdong 510080 China
                [9 ]ISNI 0000 0004 0504 5814, GRID grid.414303.1, Institute of Metabolic Disease, , Baylor Research Institute, ; 3500 Gaston Avenue, Dallas, TX 75246 USA
                Author information
                http://orcid.org/0000-0002-6854-6195
                Article
                526
                10.1186/s13045-017-0526-8
                5655880
                29065888
                5878f08d-e7e1-49ec-9424-3ac3cd5c7f9b
                © The Author(s). 2017

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

                History
                : 30 June 2017
                : 19 September 2017
                Funding
                Funded by: NIH
                Award ID: HL131460-01
                Award Recipient :
                Funded by: National Natural Science Foundation of China
                Award ID: 81560051, 81260023
                Funded by: National Key R&D Program in the Twelfth Five-year Plan
                Award ID: 2014ZX09303305
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001809, National Natural Science Foundation of China;
                Award ID: 81370371, 81570394
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002338, Ministry of Education of the People's Republic of China;
                Award ID: B13037
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2017

                Oncology & Radiotherapy
                nuclear receptors (nrs),homeostasis-associated molecular pattern receptors,atherosclerosis,metabolic disease,cardiovascular disease

                Comments

                Comment on this article