Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2017

Open Access 01-12-2017 | Research

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

Authors: Luqiao Wang, Gayani Nanayakkara, Qian Yang, Hongmei Tan, Charles Drummer, Yu Sun, Ying Shao, Hangfei Fu, Ramon Cueto, Huimin Shan, Teodoro Bottiglieri, Ya-feng Li, Candice Johnson, William Y. Yang, Fan Yang, Yanjie Xu, Hang Xi, Weiqing Liu, Jun Yu, Eric T. Choi, Xiaoshu Cheng, Hong Wang, Xiaofeng Yang

Published in: Journal of Hematology & Oncology | Issue 1/2017

Login to get access

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.
Appendix
Available only for authorised users
Literature
1.
go back to reference Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for PAMPs and suppressed by regulatory t cells. Drug Discov Today Ther Strateg. 2008;5(2):125–42.PubMedPubMedCentralCrossRef Yang XF, Yin Y, Wang H. Vascular inflammation and atherogenesis are activated via receptors for PAMPs and suppressed by regulatory t cells. Drug Discov Today Ther Strateg. 2008;5(2):125–42.PubMedPubMedCentralCrossRef
2.
go back to reference Yin Y, Yan Y, Jiang X, Mai J, Chen NC, Wang H, Yang XF. Inflammasomes are differentially expressed in cardiovascular and other tissues. Int J Immunopathol Pharmacol. 2009;22(2):311–22.PubMedPubMedCentralCrossRef Yin Y, Yan Y, Jiang X, Mai J, Chen NC, Wang H, Yang XF. Inflammasomes are differentially expressed in cardiovascular and other tissues. Int J Immunopathol Pharmacol. 2009;22(2):311–22.PubMedPubMedCentralCrossRef
3.
go back to reference Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci. 2013;18:638–49.CrossRef Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci. 2013;18:638–49.CrossRef
5.
go back to reference Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, Yang WY, Li X, Cueto R, Yu J, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation—novel paradigm and therapeutic potential. J Cardiovasc Transl Res. 2016;9(4):343–59.PubMedPubMedCentralCrossRef Wang X, Li YF, Nanayakkara G, Shao Y, Liang B, Cole L, Yang WY, Li X, Cueto R, Yu J, et al. Lysophospholipid receptors, as novel conditional danger receptors and homeostatic receptors modulate inflammation—novel paradigm and therapeutic potential. J Cardiovasc Transl Res. 2016;9(4):343–59.PubMedPubMedCentralCrossRef
6.
go back to reference Shen J, Yin Y, Mai J, Xiong X, Pansuria M, Liu J, Maley E, Saqib NU, Wang H, Yang XF. Caspase-1 recognizes extended cleavage sites in its natural substrates. Atherosclerosis. 2010;210(2):422–9.PubMedCrossRef Shen J, Yin Y, Mai J, Xiong X, Pansuria M, Liu J, Maley E, Saqib NU, Wang H, Yang XF. Caspase-1 recognizes extended cleavage sites in its natural substrates. Atherosclerosis. 2010;210(2):422–9.PubMedCrossRef
7.
go back to reference Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction- a novel mechanism for maintaining vascular function. J Hematol Oncol. 2014;7(1):80.PubMedPubMedCentralCrossRef Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction- a novel mechanism for maintaining vascular function. J Hematol Oncol. 2014;7(1):80.PubMedPubMedCentralCrossRef
8.
go back to reference Yin Y, Li X, Sha X, Xi H, Li YF, Shao Y, Mai J, Virtue A, Lopez-Pastrana J, Meng S, et al. Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arterioscler Thromb Vasc Biol. 2015;35(4):804–16.PubMedPubMedCentralCrossRef Yin Y, Li X, Sha X, Xi H, Li YF, Shao Y, Mai J, Virtue A, Lopez-Pastrana J, Meng S, et al. Early hyperlipidemia promotes endothelial activation via a caspase-1-sirtuin 1 pathway. Arterioscler Thromb Vasc Biol. 2015;35(4):804–16.PubMedPubMedCentralCrossRef
9.
go back to reference Lopez-Pastrana J, Ferrer L, Li YF, Xiong X, Xi H, Cueto R, Nelson JZ, Sha X, Li X, Cannella AL, et al. Inhibition of caspase-1 activation in endothelial cells improves angiogenesis—a novel therapeutic potential for ischemia. J Biol Chem. 2015;290(28):17485–94.PubMedPubMedCentralCrossRef Lopez-Pastrana J, Ferrer L, Li YF, Xiong X, Xi H, Cueto R, Nelson JZ, Sha X, Li X, Cannella AL, et al. Inhibition of caspase-1 activation in endothelial cells improves angiogenesis—a novel therapeutic potential for ischemia. J Biol Chem. 2015;290(28):17485–94.PubMedPubMedCentralCrossRef
10.
go back to reference Li YF, Ren LN, Guo G, Cannella LA, Chernaya V, Samuel S, Liu SX, Wang H, Yang XF. Endothelial progenitor cells in ischemic stroke: an exploration from hypothesis to therapy. J Hematol Oncol. 2015;8(1):33.PubMedPubMedCentralCrossRef Li YF, Ren LN, Guo G, Cannella LA, Chernaya V, Samuel S, Liu SX, Wang H, Yang XF. Endothelial progenitor cells in ischemic stroke: an exploration from hypothesis to therapy. J Hematol Oncol. 2015;8(1):33.PubMedPubMedCentralCrossRef
11.
go back to reference Li Y-F, Huang X, Li X, Gong R, Yin Y, Nelson J, Gao E, Zhang H, Hoffman NE, Houser SR, Madesh M, Tilley DG, Choi ET, Jiang X, Huang C-X, Wang H, Yang X-F. Caspase-1 mediates hyperlipidemia-weakened progenitor cell vessel repair. Front Biosci (Landmark Ed). 2016;21:178–91.CrossRef Li Y-F, Huang X, Li X, Gong R, Yin Y, Nelson J, Gao E, Zhang H, Hoffman NE, Houser SR, Madesh M, Tilley DG, Choi ET, Jiang X, Huang C-X, Wang H, Yang X-F. Caspase-1 mediates hyperlipidemia-weakened progenitor cell vessel repair. Front Biosci (Landmark Ed). 2016;21:178–91.CrossRef
12.
go back to reference Ferrer LM, Monroy AM, Lopez-Pastrana J, Nanayakkara G, Cueto R, Li YF, Li X, Wang H, Yang XF, Choi ET. Caspase-1 plays a critical role in accelerating chronic kidney disease-promoted neointimal hyperplasia in the carotid artery. J Cardiovasc Transl Res. 2016;9(2):135–44.PubMedPubMedCentralCrossRef Ferrer LM, Monroy AM, Lopez-Pastrana J, Nanayakkara G, Cueto R, Li YF, Li X, Wang H, Yang XF, Choi ET. Caspase-1 plays a critical role in accelerating chronic kidney disease-promoted neointimal hyperplasia in the carotid artery. J Cardiovasc Transl Res. 2016;9(2):135–44.PubMedPubMedCentralCrossRef
13.
go back to reference Xi H, Zhang Y, Xu Y, Yang WY, Jiang X, Sha X, Cheng X, Wang J, Qin X, Yu J, et al. Caspase-1 inflammasome activation mediates homocysteine-induced pyrop-apoptosis in endothelial cells. Circ Res. 2016;118(10):1525–39.PubMedPubMedCentralCrossRef Xi H, Zhang Y, Xu Y, Yang WY, Jiang X, Sha X, Cheng X, Wang J, Qin X, Yu J, et al. Caspase-1 inflammasome activation mediates homocysteine-induced pyrop-apoptosis in endothelial cells. Circ Res. 2016;118(10):1525–39.PubMedPubMedCentralCrossRef
14.
go back to reference Wang L, Fu H, Nanayakkara G, Li Y, Shao Y, Johnson C, Cheng J, Yang WY, Yang F, Lavallee M, et al. Novel extracellular and nuclear caspase-1 and inflammasomes propagate inflammation and regulate gene expression: a comprehensive database mining study. J Hematol Oncol. 2016;9(1):122.PubMedPubMedCentralCrossRef Wang L, Fu H, Nanayakkara G, Li Y, Shao Y, Johnson C, Cheng J, Yang WY, Yang F, Lavallee M, et al. Novel extracellular and nuclear caspase-1 and inflammasomes propagate inflammation and regulate gene expression: a comprehensive database mining study. J Hematol Oncol. 2016;9(1):122.PubMedPubMedCentralCrossRef
15.
go back to reference Li YF, Nanayakkara G, Sun Y, Li X, Wang L, Cueto R, Shao Y, Fu H, Johnson C, Cheng J, et al. Analyses of caspase-1-regulated transcriptomes in various tissues lead to identification of novel IL-1beta-, IL-18- and sirtuin-1-independent pathways. J Hematol Oncol. 2017;10(1):40.PubMedPubMedCentralCrossRef Li YF, Nanayakkara G, Sun Y, Li X, Wang L, Cueto R, Shao Y, Fu H, Johnson C, Cheng J, et al. Analyses of caspase-1-regulated transcriptomes in various tissues lead to identification of novel IL-1beta-, IL-18- and sirtuin-1-independent pathways. J Hematol Oncol. 2017;10(1):40.PubMedPubMedCentralCrossRef
16.
go back to reference Zhang Z, Burch PE, Cooney AJ, Lanz RB, Pereira FA, Wu J, Gibbs RA, Weinstock G, Wheeler DA. Genomic analysis of the nuclear receptor family: new insights into structure, regulation, and evolution from the rat genome. Genome Res. 2004;14(4):580–90.PubMedPubMedCentralCrossRef Zhang Z, Burch PE, Cooney AJ, Lanz RB, Pereira FA, Wu J, Gibbs RA, Weinstock G, Wheeler DA. Genomic analysis of the nuclear receptor family: new insights into structure, regulation, and evolution from the rat genome. Genome Res. 2004;14(4):580–90.PubMedPubMedCentralCrossRef
17.
go back to reference A unified nomenclature system for the nuclear receptor superfamily. Cell. 1999;97(2):161–3. A unified nomenclature system for the nuclear receptor superfamily. Cell. 1999;97(2):161–3.
18.
go back to reference Laudet V. Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor. J Mol Endocrinol. 1997;19(3):207–26.PubMedCrossRef Laudet V. Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor. J Mol Endocrinol. 1997;19(3):207–26.PubMedCrossRef
19.
21.
go back to reference Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: opening the X-files. Science. 2001;294(5548):1866–70.PubMedCrossRef Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: opening the X-files. Science. 2001;294(5548):1866–70.PubMedCrossRef
22.
go back to reference Kliewer SA, Xu HE, Lambert MH, Willson TM. Peroxisome proliferator-activated receptors: from genes to physiology. Recent Prog Horm Res. 2001;56:239–63.PubMedCrossRef Kliewer SA, Xu HE, Lambert MH, Willson TM. Peroxisome proliferator-activated receptors: from genes to physiology. Recent Prog Horm Res. 2001;56:239–63.PubMedCrossRef
23.
go back to reference Sonoda J, Pei L, Evans RM. Nuclear receptors: decoding metabolic disease. FEBS Lett. 2008;582(1):2–9.PubMedCrossRef Sonoda J, Pei L, Evans RM. Nuclear receptors: decoding metabolic disease. FEBS Lett. 2008;582(1):2–9.PubMedCrossRef
24.
go back to reference Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, Meng S, Arning E, Bottiglieri T, Choi ET, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes. 2014;63(12):4275–90. Fang P, Zhang D, Cheng Z, Yan C, Jiang X, Kruger WD, Meng S, Arning E, Bottiglieri T, Choi ET, et al. Hyperhomocysteinemia potentiates hyperglycemia-induced inflammatory monocyte differentiation and atherosclerosis. Diabetes. 2014;63(12):4275–90.
25.
26.
go back to reference Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell. 2006;126(4):789–99.PubMedCrossRef Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell. 2006;126(4):789–99.PubMedCrossRef
27.
go back to reference Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M, Mangelsdorf DJ, Evans RM. Nuclear receptor expression links the circadian clock to metabolism. Cell. 2006;126(4):801–10.PubMedCrossRef Yang X, Downes M, Yu RT, Bookout AL, He W, Straume M, Mangelsdorf DJ, Evans RM. Nuclear receptor expression links the circadian clock to metabolism. Cell. 2006;126(4):801–10.PubMedCrossRef
28.
go back to reference Lee KS, Park SJ, Hwang PH, Yi HK, Song CH, Chai OH, Kim JS, Lee MK, Lee YC. PPAR-gamma modulates allergic inflammation through up-regulation of PTEN. FASEB J. 2005;19(8):1033–5.PubMed Lee KS, Park SJ, Hwang PH, Yi HK, Song CH, Chai OH, Kim JS, Lee MK, Lee YC. PPAR-gamma modulates allergic inflammation through up-regulation of PTEN. FASEB J. 2005;19(8):1033–5.PubMed
29.
go back to reference Perez-Schindler J, Philp A. Regulation of skeletal muscle mitochondrial function by nuclear receptors: implications for health and disease. Clin Sci (Lond). 2015;129(7):589–99.CrossRef Perez-Schindler J, Philp A. Regulation of skeletal muscle mitochondrial function by nuclear receptors: implications for health and disease. Clin Sci (Lond). 2015;129(7):589–99.CrossRef
30.
go back to reference Forthmann B, Aletta JM, Lee YW, Terranova C, Birkaya B, Stachowiak EK, Stachowiak MK, Claus P. Coalition of nuclear receptors in the nervous system. J Cell Physiol. 2015;230(12):2875–80.PubMedCrossRef Forthmann B, Aletta JM, Lee YW, Terranova C, Birkaya B, Stachowiak EK, Stachowiak MK, Claus P. Coalition of nuclear receptors in the nervous system. J Cell Physiol. 2015;230(12):2875–80.PubMedCrossRef
31.
go back to reference Abergel Z, Chatterjee AK, Zuckerman B, Gross E. Regulation of neuronal oxygen responses in C. elegans is mediated through interactions between globin 5 and the H-NOX domains of soluble guanylate cyclases. J Neurosci. 2016;36(3):963–78.PubMedPubMedCentralCrossRef Abergel Z, Chatterjee AK, Zuckerman B, Gross E. Regulation of neuronal oxygen responses in C. elegans is mediated through interactions between globin 5 and the H-NOX domains of soluble guanylate cyclases. J Neurosci. 2016;36(3):963–78.PubMedPubMedCentralCrossRef
32.
go back to reference Chen NC, Yang F, Capecci LM, Gu Z, Schafer AI, Durante W, Yang XF, Wang H. Regulation of homocysteine metabolism and methylation in human and mouse tissues. FASEB J. 2010;24(8):2804–17.PubMedPubMedCentralCrossRef Chen NC, Yang F, Capecci LM, Gu Z, Schafer AI, Durante W, Yang XF, Wang H. Regulation of homocysteine metabolism and methylation in human and mouse tissues. FASEB J. 2010;24(8):2804–17.PubMedPubMedCentralCrossRef
33.
go back to reference Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.PubMedCrossRef Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–76.PubMedCrossRef
34.
go back to reference Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci (Landmark Ed). 2013;18:638–49.CrossRef Yin Y, Pastrana JL, Li X, Huang X, Mallilankaraman K, Choi ET, Madesh M, Wang H, Yang XF. Inflammasomes: sensors of metabolic stresses for vascular inflammation. Front Biosci (Landmark Ed). 2013;18:638–49.CrossRef
36.
go back to reference Choubey D, Panchanathan R. IFI16, an amplifier of DNA-damage response: role in cellular senescence and aging-associated inflammatory diseases. Ageing Res Rev. 2016;28:27–36.PubMedCrossRef Choubey D, Panchanathan R. IFI16, an amplifier of DNA-damage response: role in cellular senescence and aging-associated inflammatory diseases. Ageing Res Rev. 2016;28:27–36.PubMedCrossRef
37.
go back to reference Gurung P, Kanneganti TD. Immune responses against protozoan parasites: a focus on the emerging role of Nod-like receptors. Cell Mol Life Sci. 2016;73(16):3035–51.PubMedPubMedCentralCrossRef Gurung P, Kanneganti TD. Immune responses against protozoan parasites: a focus on the emerging role of Nod-like receptors. Cell Mol Life Sci. 2016;73(16):3035–51.PubMedPubMedCentralCrossRef
38.
go back to reference Yang J, Liu Z, Xiao TS. Post-translational regulation of inflammasomes. Cell Mol Immunol. 2017;14(1):65–79.PubMedCrossRef Yang J, Liu Z, Xiao TS. Post-translational regulation of inflammasomes. Cell Mol Immunol. 2017;14(1):65–79.PubMedCrossRef
41.
go back to reference Yang J, Fang P, Yu D, Zhang L, Zhang D, Jiang X, Yang WY, Bottiglieri T, Kunapuli SP, Yu J, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res. 2016;119(11):1226–41.PubMedCrossRef Yang J, Fang P, Yu D, Zhang L, Zhang D, Jiang X, Yang WY, Bottiglieri T, Kunapuli SP, Yu J, et al. Chronic kidney disease induces inflammatory CD40+ monocyte differentiation via homocysteine elevation and DNA hypomethylation. Circ Res. 2016;119(11):1226–41.PubMedCrossRef
42.
43.
go back to reference Medici V, Schroeder DI, Woods R, LaSalle JM, Geng Y, Shibata NM, Peerson J, Hodzic E, Dayal S, Tsukamoto H, et al. Methylation and gene expression responses to ethanol feeding and betaine supplementation in the cystathionine beta synthase-deficient mouse. Alcohol Clin Exp Res. 2014;38(6):1540–9.PubMedPubMedCentralCrossRef Medici V, Schroeder DI, Woods R, LaSalle JM, Geng Y, Shibata NM, Peerson J, Hodzic E, Dayal S, Tsukamoto H, et al. Methylation and gene expression responses to ethanol feeding and betaine supplementation in the cystathionine beta synthase-deficient mouse. Alcohol Clin Exp Res. 2014;38(6):1540–9.PubMedPubMedCentralCrossRef
44.
go back to reference Ottaviano YL, Issa JP, Parl FF, Smith HS, Baylin SB, Davidson NE. Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer cells. Cancer Res. 1994;54(10):2552–5.PubMed Ottaviano YL, Issa JP, Parl FF, Smith HS, Baylin SB, Davidson NE. Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer cells. Cancer Res. 1994;54(10):2552–5.PubMed
45.
go back to reference Issa JP, Zehnbauer BA, Civin CI, Collector MI, Sharkis SJ, Davidson NE, Kaufmann SH, Baylin SB. The estrogen receptor CpG island is methylated in most hematopoietic neoplasms. Cancer Res. 1996;56(5):973–7.PubMed Issa JP, Zehnbauer BA, Civin CI, Collector MI, Sharkis SJ, Davidson NE, Kaufmann SH, Baylin SB. The estrogen receptor CpG island is methylated in most hematopoietic neoplasms. Cancer Res. 1996;56(5):973–7.PubMed
46.
go back to reference Issa JP, Baylin SB, Belinsky SA. Methylation of the estrogen receptor CpG island in lung tumors is related to the specific type of carcinogen exposure. Cancer Res. 1996;56(16):3655–8.PubMed Issa JP, Baylin SB, Belinsky SA. Methylation of the estrogen receptor CpG island in lung tumors is related to the specific type of carcinogen exposure. Cancer Res. 1996;56(16):3655–8.PubMed
47.
go back to reference Post WS, Goldschmidt-Clermont PJ, Wilhide CC, Heldman AW, Sussman MS, Ouyang P, Milliken EE, Issa JP. Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res. 1999;43(4):985–91.PubMedCrossRef Post WS, Goldschmidt-Clermont PJ, Wilhide CC, Heldman AW, Sussman MS, Ouyang P, Milliken EE, Issa JP. Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res. 1999;43(4):985–91.PubMedCrossRef
48.
go back to reference Sasaki M, Tanaka Y, Perinchery G, Dharia A, Kotcherguina I, Fujimoto S, Dahiya R. Methylation and inactivation of estrogen, progesterone, and androgen receptors in prostate cancer. J Natl Cancer Inst. 2002;94(5):384–90.PubMedCrossRef Sasaki M, Tanaka Y, Perinchery G, Dharia A, Kotcherguina I, Fujimoto S, Dahiya R. Methylation and inactivation of estrogen, progesterone, and androgen receptors in prostate cancer. J Natl Cancer Inst. 2002;94(5):384–90.PubMedCrossRef
49.
go back to reference Lapidus RG, Nass SJ, Butash KA, Parl FF, Weitzman SA, Graff JG, Herman JG, Davidson NE. Mapping of ER gene CpG island methylation-specific polymerase chain reaction. Cancer Res. 1998;58(12):2515–9.PubMed Lapidus RG, Nass SJ, Butash KA, Parl FF, Weitzman SA, Graff JG, Herman JG, Davidson NE. Mapping of ER gene CpG island methylation-specific polymerase chain reaction. Cancer Res. 1998;58(12):2515–9.PubMed
50.
go back to reference Aithal GP, Grove JI. Genome-wide association studies in drug-induced liver injury: step change in understanding the pathogenesis. Semin Liver Dis. 2015;35(4):421–31.PubMedCrossRef Aithal GP, Grove JI. Genome-wide association studies in drug-induced liver injury: step change in understanding the pathogenesis. Semin Liver Dis. 2015;35(4):421–31.PubMedCrossRef
51.
go back to reference Wang L, Matsushita T, Madireddy L, Mousavi P, Baranzini SE. PINBPA: cytoscape app for network analysis of GWAS data. Bioinformatics. 2015;31(2):262–4.PubMedCrossRef Wang L, Matsushita T, Madireddy L, Mousavi P, Baranzini SE. PINBPA: cytoscape app for network analysis of GWAS data. Bioinformatics. 2015;31(2):262–4.PubMedCrossRef
52.
go back to reference Racke MK, Gocke AR, Muir M, Diab A, Drew PD, Lovett-Racke AE. Nuclear receptors and autoimmune disease: the potential of PPAR agonists to treat multiple sclerosis. J Nutr. 2006;136(3):700–3.PubMedPubMedCentral Racke MK, Gocke AR, Muir M, Diab A, Drew PD, Lovett-Racke AE. Nuclear receptors and autoimmune disease: the potential of PPAR agonists to treat multiple sclerosis. J Nutr. 2006;136(3):700–3.PubMedPubMedCentral
53.
go back to reference Shirinsky IV, Shirinsky VS. Targeting nuclear hormone receptors: PPARalpha agonists as potential disease-modifying drugs for rheumatoid arthritis. Int J Rheumatol. 2011;2011:937843.PubMedPubMedCentralCrossRef Shirinsky IV, Shirinsky VS. Targeting nuclear hormone receptors: PPARalpha agonists as potential disease-modifying drugs for rheumatoid arthritis. Int J Rheumatol. 2011;2011:937843.PubMedPubMedCentralCrossRef
54.
go back to reference Burris TP, Busby SA, Griffin PR. Targeting orphan nuclear receptors for treatment of metabolic diseases and autoimmunity. Chem Biol. 2012;19(1):51–9.PubMedPubMedCentralCrossRef Burris TP, Busby SA, Griffin PR. Targeting orphan nuclear receptors for treatment of metabolic diseases and autoimmunity. Chem Biol. 2012;19(1):51–9.PubMedPubMedCentralCrossRef
55.
go back to reference Park BV, Pan F. The role of nuclear receptors in regulation of Th17/Treg biology and its implications for diseases. Cell Mol Immunol. 2015;12(5):533–42.PubMedPubMedCentralCrossRef Park BV, Pan F. The role of nuclear receptors in regulation of Th17/Treg biology and its implications for diseases. Cell Mol Immunol. 2015;12(5):533–42.PubMedPubMedCentralCrossRef
56.
go back to reference Takeuchi H, Yokota-Nakatsuma A, Ohoka Y, Kagechika H, Kato C, Song SY, Iwata M. Retinoid X receptor agonists modulate Foxp3(+) regulatory T cell and Th17 cell differentiation with differential dependence on retinoic acid receptor activation. J Immunol. 2013;191(7):3725–33.PubMedCrossRef Takeuchi H, Yokota-Nakatsuma A, Ohoka Y, Kagechika H, Kato C, Song SY, Iwata M. Retinoid X receptor agonists modulate Foxp3(+) regulatory T cell and Th17 cell differentiation with differential dependence on retinoic acid receptor activation. J Immunol. 2013;191(7):3725–33.PubMedCrossRef
57.
go back to reference Li AC, Palinski W. Peroxisome proliferator-activated receptors: how their effects on macrophages can lead to the development of a new drug therapy against atherosclerosis. Annu Rev Pharmacol Toxicol. 2006;46:1–39.PubMedCrossRef Li AC, Palinski W. Peroxisome proliferator-activated receptors: how their effects on macrophages can lead to the development of a new drug therapy against atherosclerosis. Annu Rev Pharmacol Toxicol. 2006;46:1–39.PubMedCrossRef
58.
go back to reference Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest. 2006;116(3):571–80.PubMedPubMedCentralCrossRef Lefebvre P, Chinetti G, Fruchart JC, Staels B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J Clin Invest. 2006;116(3):571–80.PubMedPubMedCentralCrossRef
59.
go back to reference Shahin D, Toraby EE, Abdel-Malek H, Boshra V, Elsamanoudy AZ, Shaheen D. Effect of peroxisome proliferator-activated receptor gamma agonist (pioglitazone) and methotrexate on disease activity in rheumatoid arthritis (experimental and clinical study). Clin Med Insights Arthritis Musculoskelet Disord. 2011;4:1–10.PubMedPubMedCentralCrossRef Shahin D, Toraby EE, Abdel-Malek H, Boshra V, Elsamanoudy AZ, Shaheen D. Effect of peroxisome proliferator-activated receptor gamma agonist (pioglitazone) and methotrexate on disease activity in rheumatoid arthritis (experimental and clinical study). Clin Med Insights Arthritis Musculoskelet Disord. 2011;4:1–10.PubMedPubMedCentralCrossRef
60.
go back to reference Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol. 2017;10(1):89.PubMedPubMedCentralCrossRef Knutson TP, Truong TH, Ma S, Brady NJ, Sullivan ME, Raj G, Schwertfeger KL, Lange CA. Posttranslationally modified progesterone receptors direct ligand-specific expression of breast cancer stem cell-associated gene programs. J Hematol Oncol. 2017;10(1):89.PubMedPubMedCentralCrossRef
62.
go back to reference Sartor AO. Progression of metastatic castrate-resistant prostate cancer: impact of therapeutic intervention in the post-docetaxel space. J Hematol Oncol. 2011;4:18.PubMedPubMedCentralCrossRef Sartor AO. Progression of metastatic castrate-resistant prostate cancer: impact of therapeutic intervention in the post-docetaxel space. J Hematol Oncol. 2011;4:18.PubMedPubMedCentralCrossRef
63.
go back to reference Ross-Innes CS, Stark R, Holmes KA, Schmidt D, Spyrou C, Russell R, Massie CE, Vowler SL, Eldridge M, Carroll JS. Cooperative interaction between retinoic acid receptor-alpha and estrogen receptor in breast cancer. Genes Dev. 2010;24(2):171–82.PubMedPubMedCentralCrossRef Ross-Innes CS, Stark R, Holmes KA, Schmidt D, Spyrou C, Russell R, Massie CE, Vowler SL, Eldridge M, Carroll JS. Cooperative interaction between retinoic acid receptor-alpha and estrogen receptor in breast cancer. Genes Dev. 2010;24(2):171–82.PubMedPubMedCentralCrossRef
64.
go back to reference Savic D, Ramaker RC, Roberts BS, Dean EC, Burwell TC, Meadows SK, Cooper SJ, Garabedian MJ, Gertz J, Myers RM. Distinct gene regulatory programs define the inhibitory effects of liver X receptors and PPARG on cancer cell proliferation. Genome Med. 2016;8(1):74.PubMedPubMedCentralCrossRef Savic D, Ramaker RC, Roberts BS, Dean EC, Burwell TC, Meadows SK, Cooper SJ, Garabedian MJ, Gertz J, Myers RM. Distinct gene regulatory programs define the inhibitory effects of liver X receptors and PPARG on cancer cell proliferation. Genome Med. 2016;8(1):74.PubMedPubMedCentralCrossRef
65.
go back to reference Meng S, Ciment S, Jan M, Tran T, Pham H, Cueto R, Yang XF, Wang H. Homocysteine induces inflammatory transcriptional signaling in monocytes. Front Biosci (Landmark Ed). 2013;18:685–95.CrossRef Meng S, Ciment S, Jan M, Tran T, Pham H, Cueto R, Yang XF, Wang H. Homocysteine induces inflammatory transcriptional signaling in monocytes. Front Biosci (Landmark Ed). 2013;18:685–95.CrossRef
67.
go back to reference Cheng Z, Elmes M, Kirkup SE, Abayasekara DR, Wathes DC. Alteration of prostaglandin production and agonist responsiveness by n-6 polyunsaturated fatty acids in endometrial cells from late-gestation ewes. J Endocrinol. 2004;182(2):249–56.PubMedCrossRef Cheng Z, Elmes M, Kirkup SE, Abayasekara DR, Wathes DC. Alteration of prostaglandin production and agonist responsiveness by n-6 polyunsaturated fatty acids in endometrial cells from late-gestation ewes. J Endocrinol. 2004;182(2):249–56.PubMedCrossRef
68.
go back to reference Yang XF, Mirkovic D, Zhang S, Zhang QE, Yan Y, Xiong Z, Yang F, Chen IH, Li L, Wang H. Processing sites are different in the generation of HLA-A2.1-restricted, T cell reactive tumor antigen epitopes and viral epitopes. Int J Immunopathol Pharmacol. 2006;19(4):853–70.PubMedPubMedCentralCrossRef Yang XF, Mirkovic D, Zhang S, Zhang QE, Yan Y, Xiong Z, Yang F, Chen IH, Li L, Wang H. Processing sites are different in the generation of HLA-A2.1-restricted, T cell reactive tumor antigen epitopes and viral epitopes. Int J Immunopathol Pharmacol. 2006;19(4):853–70.PubMedPubMedCentralCrossRef
69.
go back to reference Li X, Mai J, Virtue A, Yin Y, Gong R, Sha X, Gutchigian S, Frisch A, Hodge I, Jiang X, et al. IL-35 is a novel responsive anti-inflammatory cytokine—a new system of categorizing anti-inflammatory cytokines. PLoS One. 2012;7(3):e33628.PubMedPubMedCentralCrossRef Li X, Mai J, Virtue A, Yin Y, Gong R, Sha X, Gutchigian S, Frisch A, Hodge I, Jiang X, et al. IL-35 is a novel responsive anti-inflammatory cytokine—a new system of categorizing anti-inflammatory cytokines. PLoS One. 2012;7(3):e33628.PubMedPubMedCentralCrossRef
70.
go back to reference Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997;138(3):863–70.PubMedCrossRef Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997;138(3):863–70.PubMedCrossRef
71.
go back to reference Nishimura M, Naito S, Yokoi T. Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet. 2004;19(2):135–49.PubMedCrossRef Nishimura M, Naito S, Yokoi T. Tissue-specific mRNA expression profiles of human nuclear receptor subfamilies. Drug Metab Pharmacokinet. 2004;19(2):135–49.PubMedCrossRef
72.
go back to reference D'Amore S, Vacca M, Graziano G, D'Orazio A, Cariello M, Martelli N, Di Tullio G, Salvia R, Grandaliano G, Belfiore A, et al. Nuclear receptors expression chart in peripheral blood mononuclear cells identifies patients with Metabolic Syndrome. Biochim Biophys Acta. 2013;1832(12):2289–301.PubMedCrossRef D'Amore S, Vacca M, Graziano G, D'Orazio A, Cariello M, Martelli N, Di Tullio G, Salvia R, Grandaliano G, Belfiore A, et al. Nuclear receptors expression chart in peripheral blood mononuclear cells identifies patients with Metabolic Syndrome. Biochim Biophys Acta. 2013;1832(12):2289–301.PubMedCrossRef
73.
go back to reference Kuhn TS. The structure ofscientific revolutions(3rd ed.). Chicago: University of Chicago Press; 1996.CrossRef Kuhn TS. The structure ofscientific revolutions(3rd ed.). Chicago: University of Chicago Press; 1996.CrossRef
74.
go back to reference Yang WY, Shao Y, Lopez-Pastrana J, Mai J, Wang H, Yang XF. Pathological conditions re-shape physiological Tregs into pathological Tregs. Burns Trauma. 2015;3(1). Yang WY, Shao Y, Lopez-Pastrana J, Mai J, Wang H, Yang XF. Pathological conditions re-shape physiological Tregs into pathological Tregs. Burns Trauma. 2015;3(1).
75.
go back to reference Sha X, Meng S, Li X, Xi H, Maddaloni M, Pascual DW, Shan H, Jiang X, Wang H, Yang XF. Interleukin-35 inhibits endothelial cell activation by suppressing MAPK-AP-1 pathway. J Biol Chem. 2015;290(31):19307–18.PubMedPubMedCentralCrossRef Sha X, Meng S, Li X, Xi H, Maddaloni M, Pascual DW, Shan H, Jiang X, Wang H, Yang XF. Interleukin-35 inhibits endothelial cell activation by suppressing MAPK-AP-1 pathway. J Biol Chem. 2015;290(31):19307–18.PubMedPubMedCentralCrossRef
76.
go back to reference Sun Y, Johnson C, Zhou J, Wang L, Li Y-F, Lu Y, Nanayakkara G, Fu H, Shao Y, Sanchez C, Yang WY, Wang X, Choi ET, Li R, Wang H, Yang X-F. Uremic toxins are conditional danger- or homeostasis-associated molecular patterns. Front Biosci. 2017;22(23):348–87. Sun Y, Johnson C, Zhou J, Wang L, Li Y-F, Lu Y, Nanayakkara G, Fu H, Shao Y, Sanchez C, Yang WY, Wang X, Choi ET, Li R, Wang H, Yang X-F. Uremic toxins are conditional danger- or homeostasis-associated molecular patterns. Front Biosci. 2017;22(23):348–87.
77.
go back to reference Basil MC, Levy BD. Specialized pro-resolving mediators: endogenous regulators of infection and inflammation. Nat Rev Immunol. 2016;16(1):51–67.PubMedCrossRef Basil MC, Levy BD. Specialized pro-resolving mediators: endogenous regulators of infection and inflammation. Nat Rev Immunol. 2016;16(1):51–67.PubMedCrossRef
78.
go back to reference Wallace BD, Redinbo MR. Xenobiotic-sensing nuclear receptors involved in drug metabolism: a structural perspective. Drug Metab Rev. 2013;45(1):79–100.PubMedCrossRef Wallace BD, Redinbo MR. Xenobiotic-sensing nuclear receptors involved in drug metabolism: a structural perspective. Drug Metab Rev. 2013;45(1):79–100.PubMedCrossRef
79.
go back to reference Leclercq G, Jacquot Y. Interactions of isoflavones and other plant derived estrogens with estrogen receptors for prevention and treatment of breast cancer-considerations concerning related efficacy and safety. J Steroid Biochem Mol Biol. 2014;139:237–44.PubMedCrossRef Leclercq G, Jacquot Y. Interactions of isoflavones and other plant derived estrogens with estrogen receptors for prevention and treatment of breast cancer-considerations concerning related efficacy and safety. J Steroid Biochem Mol Biol. 2014;139:237–44.PubMedCrossRef
80.
go back to reference Bloom MS, Mok-Lin E, Fujimoto VY. Bisphenol A and ovarian steroidogenesis. Fertil Steril. 2016;106(4):857–63.PubMedCrossRef Bloom MS, Mok-Lin E, Fujimoto VY. Bisphenol A and ovarian steroidogenesis. Fertil Steril. 2016;106(4):857–63.PubMedCrossRef
81.
go back to reference Eick GN, Colucci JK, Harms MJ, Ortlund EA, Thornton JW. Evolution of minimal specificity and promiscuity in steroid hormone receptors. PLoS Genet. 2012;8(11):e1003072.PubMedPubMedCentralCrossRef Eick GN, Colucci JK, Harms MJ, Ortlund EA, Thornton JW. Evolution of minimal specificity and promiscuity in steroid hormone receptors. PLoS Genet. 2012;8(11):e1003072.PubMedPubMedCentralCrossRef
82.
go back to reference Yoshikuni Y, Ferrin TE, Keasling JD. Designed divergent evolution of enzyme function. Nature. 2006;440(7087):1078–82.PubMedCrossRef Yoshikuni Y, Ferrin TE, Keasling JD. Designed divergent evolution of enzyme function. Nature. 2006;440(7087):1078–82.PubMedCrossRef
83.
84.
go back to reference O'Brien PJ, Herschlag D. Catalytic promiscuity and the evolution of new enzymatic activities. Chem Biol. 1999;6(4):R91–R105.PubMedCrossRef O'Brien PJ, Herschlag D. Catalytic promiscuity and the evolution of new enzymatic activities. Chem Biol. 1999;6(4):R91–R105.PubMedCrossRef
85.
go back to reference Noy N. Ligand specificity of nuclear hormone receptors: sifting through promiscuity. Biochemistry. 2007;46(47):13461–7.PubMedCrossRef Noy N. Ligand specificity of nuclear hormone receptors: sifting through promiscuity. Biochemistry. 2007;46(47):13461–7.PubMedCrossRef
86.
go back to reference Ortlund EA, Lee Y, Solomon IH, Hager JM, Safi R, Choi Y, Guan Z, Tripathy A, Raetz CR, McDonnell DP, et al. Modulation of human nuclear receptor LRH-1 activity by phospholipids and SHP. Nat Struct Mol Biol. 2005;12(4):357–63.PubMedCrossRef Ortlund EA, Lee Y, Solomon IH, Hager JM, Safi R, Choi Y, Guan Z, Tripathy A, Raetz CR, McDonnell DP, et al. Modulation of human nuclear receptor LRH-1 activity by phospholipids and SHP. Nat Struct Mol Biol. 2005;12(4):357–63.PubMedCrossRef
87.
go back to reference Bourguet W, Vivat V, Wurtz JM, Chambon P, Gronemeyer H, Moras D. Crystal structure of a heterodimeric complex of RAR and RXR ligand-binding domains. Mol Cell. 2000;5(2):289–98.PubMedCrossRef Bourguet W, Vivat V, Wurtz JM, Chambon P, Gronemeyer H, Moras D. Crystal structure of a heterodimeric complex of RAR and RXR ligand-binding domains. Mol Cell. 2000;5(2):289–98.PubMedCrossRef
88.
go back to reference Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, Evans RM, Haussler MR, Mangelsdorf DJ. Vitamin D receptor as an intestinal bile acid sensor. Science. 2002;296(5571):1313–6.PubMedCrossRef Makishima M, Lu TT, Xie W, Whitfield GK, Domoto H, Evans RM, Haussler MR, Mangelsdorf DJ. Vitamin D receptor as an intestinal bile acid sensor. Science. 2002;296(5571):1313–6.PubMedCrossRef
89.
go back to reference Levin AA, Sturzenbecker LJ, Kazmer S, Bosakowski T, Huselton C, Allenby G, Speck J, Kratzeisen C, Rosenberger M, Lovey A, et al. 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXR alpha. Nature. 1992;355(6358):359–61.PubMedCrossRef Levin AA, Sturzenbecker LJ, Kazmer S, Bosakowski T, Huselton C, Allenby G, Speck J, Kratzeisen C, Rosenberger M, Lovey A, et al. 9-cis retinoic acid stereoisomer binds and activates the nuclear receptor RXR alpha. Nature. 1992;355(6358):359–61.PubMedCrossRef
90.
go back to reference Heyman RA, Mangelsdorf DJ, Dyck JA, Stein RB, Eichele G, Evans RM, Thaller C. 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell. 1992;68(2):397–406.PubMedCrossRef Heyman RA, Mangelsdorf DJ, Dyck JA, Stein RB, Eichele G, Evans RM, Thaller C. 9-cis retinoic acid is a high affinity ligand for the retinoid X receptor. Cell. 1992;68(2):397–406.PubMedCrossRef
91.
go back to reference Shi H, Kichaev G, Pasaniuc B. Contrasting the genetic architecture of 30 complex traits from summary association data. Am J Hum Genet. 2016;99(1):139–53.PubMedPubMedCentralCrossRef Shi H, Kichaev G, Pasaniuc B. Contrasting the genetic architecture of 30 complex traits from summary association data. Am J Hum Genet. 2016;99(1):139–53.PubMedPubMedCentralCrossRef
92.
go back to reference Boyle EA, Li YI, Pritchard JK. An expanded view of complex traits: from polygenic to omnigenic. Cell. 2017;169(7):1177–86.PubMedCrossRef Boyle EA, Li YI, Pritchard JK. An expanded view of complex traits: from polygenic to omnigenic. Cell. 2017;169(7):1177–86.PubMedCrossRef
93.
go back to reference Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, et al. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–53.PubMedPubMedCentralCrossRef Manolio TA, Collins FS, Cox NJ, Goldstein DB, Hindorff LA, Hunter DJ, McCarthy MI, Ramos EM, Cardon LR, Chakravarti A, et al. Finding the missing heritability of complex diseases. Nature. 2009;461(7265):747–53.PubMedPubMedCentralCrossRef
94.
go back to reference Purcell SM, Wray NR, Stone JL, Visscher PM, O'Donovan MC, Sullivan PF, Sklar P. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;460(7256):748–52.PubMed Purcell SM, Wray NR, Stone JL, Visscher PM, O'Donovan MC, Sullivan PF, Sklar P. Common polygenic variation contributes to risk of schizophrenia and bipolar disorder. Nature. 2009;460(7256):748–52.PubMed
95.
96.
go back to reference Chakravarti A, Turner TN. Revealing rate-limiting steps in complex disease biology: the crucial importance of studying rare, extreme-phenotype families. Bioessays. 2016;38(6):578–86.PubMedCrossRef Chakravarti A, Turner TN. Revealing rate-limiting steps in complex disease biology: the crucial importance of studying rare, extreme-phenotype families. Bioessays. 2016;38(6):578–86.PubMedCrossRef
97.
go back to reference Vassy JL, Hivert MF, Porneala B, Dauriz M, Florez JC, Dupuis J, Siscovick DS, Fornage M, Rasmussen-Torvik LJ, Bouchard C, et al. Polygenic type 2 diabetes prediction at the limit of common variant detection. Diabetes. 2014;63(6):2172–82.PubMedPubMedCentralCrossRef Vassy JL, Hivert MF, Porneala B, Dauriz M, Florez JC, Dupuis J, Siscovick DS, Fornage M, Rasmussen-Torvik LJ, Bouchard C, et al. Polygenic type 2 diabetes prediction at the limit of common variant detection. Diabetes. 2014;63(6):2172–82.PubMedPubMedCentralCrossRef
98.
go back to reference Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002;106(25):3143–421. Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation. 2002;106(25):3143–421.
99.
go back to reference Alberti KG, Zimmet P, Shaw J. The metabolic syndrome—a new worldwide definition. Lancet. 2005;366(9491):1059–62.PubMedCrossRef Alberti KG, Zimmet P, Shaw J. The metabolic syndrome—a new worldwide definition. Lancet. 2005;366(9491):1059–62.PubMedCrossRef
100.
go back to reference Mark M, Ghyselinck NB, Chambon P. Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu Rev Pharmacol Toxicol. 2006;46:451–80.PubMedCrossRef Mark M, Ghyselinck NB, Chambon P. Function of retinoid nuclear receptors: lessons from genetic and pharmacological dissections of the retinoic acid signaling pathway during mouse embryogenesis. Annu Rev Pharmacol Toxicol. 2006;46:451–80.PubMedCrossRef
101.
go back to reference Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352(16):1685–95.PubMedCrossRef Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005;352(16):1685–95.PubMedCrossRef
102.
go back to reference Hilgendorf I, Gerhardt LM, Tan TC, Winter C, Holderried TA, Chousterman BG, Iwamoto Y, Liao R, Zirlik A, Scherer-Crosbie M. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. 2014;114(10):1611–22.PubMedPubMedCentralCrossRef Hilgendorf I, Gerhardt LM, Tan TC, Winter C, Holderried TA, Chousterman BG, Iwamoto Y, Liao R, Zirlik A, Scherer-Crosbie M. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. 2014;114(10):1611–22.PubMedPubMedCentralCrossRef
103.
go back to reference Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci U S A. 2006;103(4):1006–11.PubMedPubMedCentralCrossRef Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci U S A. 2006;103(4):1006–11.PubMedPubMedCentralCrossRef
104.
go back to reference Giudici M, Goni S, Fan R, Treuter E. Nuclear receptor coregulators in metabolism and disease. 2015.CrossRef Giudici M, Goni S, Fan R, Treuter E. Nuclear receptor coregulators in metabolism and disease. 2015.CrossRef
105.
go back to reference Vuong LM, Dhahbi J, Sladek FM. SAT-273: expression of nuclear receptor HNF4a in mouse embryonic stem cells is sufficient to inhibit cell proliferation and drive differentiation. 2015. Vuong LM, Dhahbi J, Sladek FM. SAT-273: expression of nuclear receptor HNF4a in mouse embryonic stem cells is sufficient to inhibit cell proliferation and drive differentiation. 2015.
106.
go back to reference Lin S-J, Lin C-Y, Yang D-R, Izumi K, Yan E, Niu X, Chang H-C, Miyamoto H, Wang N, Li G. The differential effects of anti-diabetic thiazolidinedione on prostate cancer progression are linked to the TR4 nuclear receptor expression status. Neoplasia. 2015;17(4):339–47.PubMedPubMedCentralCrossRef Lin S-J, Lin C-Y, Yang D-R, Izumi K, Yan E, Niu X, Chang H-C, Miyamoto H, Wang N, Li G. The differential effects of anti-diabetic thiazolidinedione on prostate cancer progression are linked to the TR4 nuclear receptor expression status. Neoplasia. 2015;17(4):339–47.PubMedPubMedCentralCrossRef
107.
go back to reference Ghoneim RH, Ngo Sock ET, Lavoie J-M, Piquette-Miller M. Effect of a high-fat diet on the hepatic expression of nuclear receptors and their target genes: relevance to drug disposition. Br J Nutr. 2015;113(03):507–16.PubMedCrossRef Ghoneim RH, Ngo Sock ET, Lavoie J-M, Piquette-Miller M. Effect of a high-fat diet on the hepatic expression of nuclear receptors and their target genes: relevance to drug disposition. Br J Nutr. 2015;113(03):507–16.PubMedCrossRef
Metadata
Title
A comprehensive data mining study shows that most nuclear receptors act as newly proposed homeostasis-associated molecular pattern receptors
Authors
Luqiao Wang
Gayani Nanayakkara
Qian Yang
Hongmei Tan
Charles Drummer
Yu Sun
Ying Shao
Hangfei Fu
Ramon Cueto
Huimin Shan
Teodoro Bottiglieri
Ya-feng Li
Candice Johnson
William Y. Yang
Fan Yang
Yanjie Xu
Hang Xi
Weiqing Liu
Jun Yu
Eric T. Choi
Xiaoshu Cheng
Hong Wang
Xiaofeng Yang
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2017
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-017-0526-8

Other articles of this Issue 1/2017

Journal of Hematology & Oncology 1/2017 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine