#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Beyond the heterodimer model for mineralocorticoid and glucocorticoid receptor interactions in nuclei and at DNA


Autoři: John R. Pooley aff001;  Caroline A. Rivers aff001;  Michael T. Kilcooley aff001;  Susana N. Paul aff001;  Ayse Derya Cavga aff003;  Yvonne M. Kershaw aff001;  Serena Muratcioglu aff004;  Attila Gursoy aff003;  Ozlem Keskin aff003;  Stafford L. Lightman aff001
Působiště autorů: Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, United Kingdom aff001;  Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America aff002;  Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey aff003;  Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America aff004;  Howard Hughes Medical Institute, University of California, Berkeley, California, United States of America aff005
Vyšlo v časopise: PLoS ONE 15(1)
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pone.0227520

Souhrn

Glucocorticoid (GR) and mineralocorticoid receptors (MR) are believed to classically bind DNA as homodimers or MR-GR heterodimers to influence gene regulation in response to pulsatile basal or stress-evoked glucocorticoid secretion. Pulsed corticosterone presentation reveals MR and GR co-occupy DNA only at the peaks of glucocorticoid oscillations, allowing interaction. GR DNA occupancy was pulsatile, while MR DNA occupancy was prolonged through the inter-pulse interval. In mouse mammary 3617 cells MR-GR interacted in the nucleus and at a chromatin-associated DNA binding site. Interactions occurred irrespective of ligand type and receptors formed complexes of higher order than heterodimers. We also detected MR-GR interactions ex-vivo in rat hippocampus. An expanded range of MR-GR interactions predicts structural allostery allowing a variety of transcriptional outcomes and is applicable to the multiple tissue types that co-express both receptors in the same cells whether activated by the same or different hormones.

Klíčová slova:

DNA-binding proteins – Protein interactions – Hormones – Co-immunoprecipitation – Hippocampus – Aldosterone – Dimers – Prisms


Zdroje

1. Conway-Campbell BL, Pooley JR, Hager GL, Lightman SL. Molecular dynamics of ultradian glucocorticoid receptor action. Mol Cell Endocrinol. 2012;348(2):383–93. doi: 10.1016/j.mce.2011.08.014 21872640

2. Lightman SL, Conway-Campbell BL. The crucial role of pulsatile activity of the HPA axis for continuous dynamic equilibration. Nat Rev Neurosci. 2010;11(10):710–8. doi: 10.1038/nrn2914 20842176

3. Morsink MC, Steenbergen PJ, Vos JB, Karst H, Joels M, De Kloet ER, et al. Acute activation of hippocampal glucocorticoid receptors results in different waves of gene expression throughout time. J Neuroendocrinol. 2006;18(4):239–52. doi: 10.1111/j.1365-2826.2006.01413.x 16503919

4. Stavreva DA, Wiench M, John S, Conway-Campbell BL, McKenna MA, Pooley JR, et al. Ultradian hormone stimulation induces glucocorticoid receptor-mediated pulses of gene transcription. Nat Cell Biol. 2009;11(9):1093–102. doi: 10.1038/ncb1922 19684579

5. George CL, Birnie MT, Flynn BP, Kershaw YM, Lightman SL, Conway-Campbell BL. Ultradian glucocorticoid exposure directs gene-dependent and tissue-specific mRNA expression patterns in vivo. Mol Cell Endocrinol. 2017;439:46–53. doi: 10.1016/j.mce.2016.10.019 27769714

6. Conway-Campbell BL, Sarabdjitsingh RA, McKenna MA, Pooley JR, Kershaw YM, Meijer OC, et al. Glucocorticoid ultradian rhythmicity directs cyclical gene pulsing of the clock gene period 1 in rat hippocampus. J Neuroendocrinol. 2010;22(10):1093–100. doi: 10.1111/j.1365-2826.2010.02051.x 20649850

7. Klein JF. Adverse psychiatric effects of systemic glucocorticoid therapy. Am Fam Physician. 1992;46(5):1469–74. 1442465

8. Conway-Campbell BL, McKenna MA, Wiles CC, Atkinson HC, de Kloet ER, Lightman SL. Proteasome-dependent down-regulation of activated nuclear hippocampal glucocorticoid receptors determines dynamic responses to corticosterone. Endocrinology. 2007;148(11):5470–7. doi: 10.1210/en.2007-0585 17690167

9. Mifsud KR, Reul JM. Acute stress enhances heterodimerization and binding of corticosteroid receptors at glucocorticoid target genes in the hippocampus. Proc Natl Acad Sci U S A. 2016;113(40):11336–41. doi: 10.1073/pnas.1605246113 27655894

10. Stolte EH, van Kemenade BM, Savelkoul HF, Flik G. Evolution of glucocorticoid receptors with different glucocorticoid sensitivity. J Endocrinol. 2006;190(1):17–28. doi: 10.1677/joe.1.06703 16837607

11. Griekspoor A, Zwart W, Neefjes J, Michalides R. Visualizing the action of steroid hormone receptors in living cells. Nucl Recept Signal. 2007;5:e003. doi: 10.1621/nrs.05003 17464358

12. Luisi BF, Xu WX, Otwinowski Z, Freedman LP, Yamamoto KR, Sigler PB. Crystallographic analysis of the interaction of the glucocorticoid receptor with DNA. Nature. 1991;352(6335):497–505. doi: 10.1038/352497a0 1865905

13. Reul JM, de Kloet ER. Two receptor systems for corticosterone in rat brain: microdistribution and differential occupation. Endocrinology. 1985;117(6):2505–11. doi: 10.1210/endo-117-6-2505 2998738

14. Pearce D, Yamamoto KR. Mineralocorticoid and glucocorticoid receptor activities distinguished by nonreceptor factors at a composite response element. Science. 1993;259(5098):1161–5. doi: 10.1126/science.8382376 8382376

15. Eriksson P, Wrange O. Protein-protein contacts in the glucocorticoid receptor homodimer influence its DNA binding properties. J Biol Chem. 1990;265(6):3535–42. 2303460

16. Wrange O, Eriksson P, Perlmann T. The purified activated glucocorticoid receptor is a homodimer. J Biol Chem. 1989;264(9):5253–9. 2494184

17. Dahlman-Wright K, Siltala-Roos H, Carlstedt-Duke J, Gustafsson JA. Protein-protein interactions facilitate DNA binding by the glucocorticoid receptor DNA-binding domain. J Biol Chem. 1990;265(23):14030–5. 1974254

18. Dahlman-Wright K, Wright A, Gustafsson JA, Carlstedt-Duke J. Interaction of the glucocorticoid receptor DNA-binding domain with DNA as a dimer is mediated by a short segment of five amino acids. J Biol Chem. 1991;266(5):3107–12. 1993683

19. Drouin J, Sun YL, Tremblay S, Lavender P, Schmidt TJ, de Lean A, et al. Homodimer formation is rate-limiting for high affinity DNA binding by glucocorticoid receptor. Mol Endocrinol. 1992;6(8):1299–309. doi: 10.1210/mend.6.8.1406707 1406707

20. Fawell SE, Lees JA, White R, Parker MG. Characterization and colocalization of steroid binding and dimerization activities in the mouse estrogen receptor. Cell. 1990;60(6):953–62. doi: 10.1016/0092-8674(90)90343-d 2317866

21. Hirst MA, Hinck L, Danielsen M, Ringold GM. Discrimination of DNA response elements for thyroid hormone and estrogen is dependent on dimerization of receptor DNA binding domains. Proc Natl Acad Sci U S A. 1992;89(12):5527–31. doi: 10.1073/pnas.89.12.5527 1608965

22. Arnold SF, Notides AC. An antiestrogen: a phosphotyrosyl peptide that blocks dimerization of the human estrogen receptor. Proc Natl Acad Sci U S A. 1995;92(16):7475–9. doi: 10.1073/pnas.92.16.7475 7543683

23. Aumais JP, Lee HS, DeGannes C, Horsford J, White JH. Function of directly repeated half-sites as response elements for steroid hormone receptors. J Biol Chem. 1996;271(21):12568–77. doi: 10.1074/jbc.271.21.12568 8647867

24. Arriza JL, Weinberger C, Cerelli G, Glaser TM, Handelin BL, Housman DE, et al. Cloning of human mineralocorticoid receptor complementary DNA: structural and functional kinship with the glucocorticoid receptor. Science. 1987;237(4812):268–75. doi: 10.1126/science.3037703 3037703

25. Liu W, Wang J, Sauter NK, Pearce D. Steroid receptor heterodimerization demonstrated in vitro and in vivo. Proc Natl Acad Sci U S A. 1995;92(26):12480–4. doi: 10.1073/pnas.92.26.12480 8618925

26. Trapp T, Rupprecht R, Castren M, Reul JM, Holsboer F. Heterodimerization between mineralocorticoid and glucocorticoid receptor: a new principle of glucocorticoid action in the CNS. Neuron. 1994;13(6):1457–62. doi: 10.1016/0896-6273(94)90431-6 7993637

27. Farman N, Rafestin-Oblin ME. Multiple aspects of mineralocorticoid selectivity. Am J Physiol Renal Physiol. 2001;280(2):F181–92. doi: 10.1152/ajprenal.2001.280.2.F181 11208593

28. Kiilerich P, Triqueneaux G, Christensen NM, Trayer V, Terrien X, Lombes M, et al. Interaction between the trout mineralocorticoid and glucocorticoid receptors in vitro. J Mol Endocrinol. 2015;55(1):55–68. doi: 10.1530/JME-15-0002 26108487

29. Ou XM, Storring JM, Kushwaha N, Albert PR. Heterodimerization of mineralocorticoid and glucocorticoid receptors at a novel negative response element of the 5-HT1A receptor gene. J Biol Chem. 2001;276(17):14299–307. doi: 10.1074/jbc.M005363200 11278286

30. Derfoul A, Robertson NM, Lingrel JB, Hall DJ, Litwack G. Regulation of the human Na/K-ATPase beta1 gene promoter by mineralocorticoid and glucocorticoid receptors. J Biol Chem. 1998;273(33):20702–11. doi: 10.1074/jbc.273.33.20702 9694812

31. Planey SL, Derfoul A, Steplewski A, Robertson NM, Litwack G. Inhibition of glucocorticoid-induced apoptosis in 697 pre-B lymphocytes by the mineralocorticoid receptor N-terminal domain. J Biol Chem. 2002;277(44):42188–96. doi: 10.1074/jbc.M205085200 12194973

32. Nishi M, Tanaka M, Matsuda K, Sunaguchi M, Kawata M. Visualization of glucocorticoid receptor and mineralocorticoid receptor interactions in living cells with GFP-based fluorescence resonance energy transfer. J Neurosci. 2004;24(21):4918–27. doi: 10.1523/JNEUROSCI.5495-03.2004 15163683

33. Savory JG, Prefontaine GG, Lamprecht C, Liao M, Walther RF, Lefebvre YA, et al. Glucocorticoid receptor homodimers and glucocorticoid-mineralocorticoid receptor heterodimers form in the cytoplasm through alternative dimerization interfaces. Mol Cell Biol. 2001;21(3):781–93. doi: 10.1128/MCB.21.3.781-793.2001 11154266

34. Smith CL, Htun H, Wolford RG, Hager GL. Differential activity of progesterone and glucocorticoid receptors on mouse mammary tumor virus templates differing in chromatin structure. J Biol Chem. 1997;272(22):14227–35. doi: 10.1074/jbc.272.22.14227 9162055

35. Bledsoe RK, Montana VG, Stanley TB, Delves CJ, Apolito CJ, McKee DD, et al. Crystal structure of the glucocorticoid receptor ligand binding domain reveals a novel mode of receptor dimerization and coactivator recognition. Cell. 2002;110(1):93–105. doi: 10.1016/s0092-8674(02)00817-6 12151000

36. Liu W, Wang J, Yu G, Pearce D. Steroid receptor transcriptional synergy is potentiated by disruption of the DNA-binding domain dimer interface. Mol Endocrinol. 1996;10(11):1399–406. doi: 10.1210/mend.10.11.8923466 8923466

37. Meijsing SH, Pufall MA, So AY, Bates DL, Chen L, Yamamoto KR. DNA binding site sequence directs glucocorticoid receptor structure and activity. Science. 2009;324(5925):407–10. doi: 10.1126/science.1164265 19372434

38. Kim S, Brostromer E, Xing D, Jin J, Chong S, Ge H, et al. Probing allostery through DNA. Science. 2013;339(6121):816–9. doi: 10.1126/science.1229223 23413354

39. Lefstin JA, Yamamoto KR. Allosteric effects of DNA on transcriptional regulators. Nature. 1998;392(6679):885–8. doi: 10.1038/31860 9582068

40. Han F, Ozawa H, Matsuda K, Nishi M, Kawata M. Colocalization of mineralocorticoid receptor and glucocorticoid receptor in the hippocampus and hypothalamus. Neurosci Res. 2005;51(4):371–81. doi: 10.1016/j.neures.2004.12.013 15740800

41. Beavan S, Horner A, Bord S, Ireland D, Compston J. Colocalization of glucocorticoid and mineralocorticoid receptors in human bone. J Bone Miner Res. 2001;16(8):1496–504. doi: 10.1359/jbmr.2001.16.8.1496 11499872

42. Marzolla V, Armani A, Zennaro MC, Cinti F, Mammi C, Fabbri A, et al. The role of the mineralocorticoid receptor in adipocyte biology and fat metabolism. Mol Cell Endocrinol. 2012;350(2):281–8. doi: 10.1016/j.mce.2011.09.011 21945603

43. Armanini D, Endres S, Kuhnle U, Weber PC. Parallel determination of mineralocorticoid and glucocorticoid receptors in T- and B-lymphocytes of human spleen. Acta Endocrinol (Copenh). 1988;118(4):479–82.

44. Farman N, Oblin ME, Lombes M, Delahaye F, Westphal HM, Bonvalet JP, et al. Immunolocalization of gluco- and mineralocorticoid receptors in rabbit kidney. Am J Physiol. 1991;260(2 Pt 1):C226–33. doi: 10.1152/ajpcell.1991.260.2.C226 1847584

45. Bradbury MJ, Akana SF, Dallman MF. Roles of type I and II corticosteroid receptors in regulation of basal activity in the hypothalamo-pituitary-adrenal axis during the diurnal trough and the peak: evidence for a nonadditive effect of combined receptor occupation. Endocrinology. 1994;134(3):1286–96. doi: 10.1210/endo.134.3.8119168 8119168

46. De Kloet ER, Vreugdenhil E, Oitzl MS, Joels M. Brain corticosteroid receptor balance in health and disease. Endocr Rev. 1998;19(3):269–301. doi: 10.1210/edrv.19.3.0331 9626555

47. Joels M, de Kloet ER. Mineralocorticoid and glucocorticoid receptors in the brain. Implications for ion permeability and transmitter systems. Prog Neurobiol. 1994;43(1):1–36. doi: 10.1016/0301-0082(94)90014-0 7526416

48. McNally JG, Muller WG, Walker D, Wolford R, Hager GL. The glucocorticoid receptor: rapid exchange with regulatory sites in living cells. Science. 2000;287(5456):1262–5. doi: 10.1126/science.287.5456.1262 10678832

49. Chakraborti PK, Garabedian MJ, Yamamoto KR, Simons SS Jr. Creation of "super" glucocorticoid receptors by point mutations in the steroid binding domain. J Biol Chem. 1991;266(33):22075–8. 1939229

50. Droste SK, de Groote L, Atkinson HC, Lightman SL, Reul JM, Linthorst AC. Corticosterone levels in the brain show a distinct ultradian rhythm but a delayed response to forced swim stress. Endocrinology. 2008;149(7):3244–53. doi: 10.1210/en.2008-0103 18356272

51. Qian X, Droste SK, Lightman SL, Reul JM, Linthorst AC. Circadian and ultradian rhythms of free glucocorticoid hormone are highly synchronized between the blood, the subcutaneous tissue, and the brain. Endocrinology. 2012;153(9):4346–53. doi: 10.1210/en.2012-1484 22822164

52. Digman MA, Wiseman PW, Choi C, Horwitz AR, Gratton E. Stoichiometry of molecular complexes at adhesions in living cells. Proc Natl Acad Sci U S A. 2009;106(7):2170–5. doi: 10.1073/pnas.0806036106 19168634

53. Choi CK, Zareno J, Digman MA, Gratton E, Horwitz AR. Cross-correlated fluctuation analysis reveals phosphorylation-regulated paxillin-FAK complexes in nascent adhesions. Biophys J. 2011;100(3):583–92. doi: 10.1016/j.bpj.2010.12.3719 21281572

54. Zacharias DA, Violin JD, Newton AC, Tsien RY. Partitioning of lipid-modified monomeric GFPs into membrane microdomains of live cells. Science. 2002;296(5569):913–6. doi: 10.1126/science.1068539 11988576

55. Chen S, Wang J, Yu G, Liu W, Pearce D. Androgen and glucocorticoid receptor heterodimer formation. A possible mechanism for mutual inhibition of transcriptional activity. J Biol Chem. 1997;272(22):14087–92. doi: 10.1074/jbc.272.22.14087 9162033

56. Digman MA, Dalal R, Horwitz AF, Gratton E. Mapping the number of molecules and brightness in the laser scanning microscope. Biophys J. 2008;94(6):2320–32. doi: 10.1529/biophysj.107.114645 18096627

57. Fukushima R, Yamamoto J, Ishikawa H, Kinjo M. Two-detector number and brightness analysis reveals spatio-temporal oligomerization of proteins in living cells. Methods. 2018;140–141:161–71. doi: 10.1016/j.ymeth.2018.03.007 29572069

58. Presman DM, Ogara MF, Stortz M, Alvarez LD, Pooley JR, Schiltz RL, et al. Live cell imaging unveils multiple domain requirements for in vivo dimerization of the glucocorticoid receptor. PLoS Biol. 2014;12(3):e1001813. doi: 10.1371/journal.pbio.1001813 24642507

59. Tuncbag N, Gursoy A, Nussinov R, Keskin O. Predicting protein-protein interactions on a proteome scale by matching evolutionary and structural similarities at interfaces using PRISM. Nat Protoc. 2011;6(9):1341–54. doi: 10.1038/nprot.2011.367 21886100

60. Baspinar A, Cukuroglu E, Nussinov R, Keskin O, Gursoy A. PRISM: a web server and repository for prediction of protein-protein interactions and modeling their 3D complexes. Nucleic Acids Res. 2014;42(Web Server issue):W285–9. doi: 10.1093/nar/gku397 24829450

61. Presman DM, Ganguly S, Schiltz RL, Johnson TA, Karpova TS, Hager GL. DNA binding triggers tetramerization of the glucocorticoid receptor in live cells. Proc Natl Acad Sci U S A. 2016;113(29):8236–41. doi: 10.1073/pnas.1606774113 27382178

62. Stephens B, Handel TM. Chemokine receptor oligomerization and allostery. Prog Mol Biol Transl Sci. 2013;115:375–420. doi: 10.1016/B978-0-12-394587-7.00009-9 23415099

63. Jewell CM, Scoltock AB, Hamel BL, Yudt MR, Cidlowski JA. Complex human glucocorticoid receptor dim mutations define glucocorticoid induced apoptotic resistance in bone cells. Mol Endocrinol. 2012;26(2):244–56. doi: 10.1210/me.2011-1116 22174376

64. Rivers CA, Rogers MF, Stubbs FE, Conway-Campbell BL, Lightman SL, Pooley JR. Glucocorticoid Receptor-Tethered Mineralocorticoid Receptors Increase Glucocorticoid-Induced Transcriptional Responses. Endocrinology. 2019;160(5):1044–56. doi: 10.1210/en.2018-00819 30980716

65. Groeneweg FL, van Royen ME, Fenz S, Keizer VI, Geverts B, Prins J, et al. Quantitation of glucocorticoid receptor DNA-binding dynamics by single-molecule microscopy and FRAP. PLoS One. 2014;9(3):e90532. doi: 10.1371/journal.pone.0090532 24632838

66. Neumann FR, Dion V, Gehlen LR, Tsai-Pflugfelder M, Schmid R, Taddei A, et al. Targeted INO80 enhances subnuclear chromatin movement and ectopic homologous recombination. Genes Dev. 2012;26(4):369–83. doi: 10.1101/gad.176156.111 22345518

67. Edwards DP, Altmann M, DeMarzo A, Zhang Y, Weigel NL, Beck CA. Progesterone receptor and the mechanism of action of progesterone antagonists. J Steroid Biochem Mol Biol. 1995;53(1–6):449–58. doi: 10.1016/0960-0760(95)00091-d 7626494

68. Lehmann JM, Zhang XK, Graupner G, Lee MO, Hermann T, Hoffmann B, et al. Formation of retinoid X receptor homodimers leads to repression of T3 response: hormonal cross talk by ligand-induced squelching. Mol Cell Biol. 1993;13(12):7698–707. doi: 10.1128/mcb.13.12.7698 8246986

69. Miranda TB, Morris SA, Hager GL. Complex genomic interactions in the dynamic regulation of transcription by the glucocorticoid receptor. Mol Cell Endocrinol. 2013;380(1–2):16–24. doi: 10.1016/j.mce.2013.03.002 23499945

70. Voss TC, Schiltz RL, Sung MH, Yen PM, Stamatoyannopoulos JA, Biddie SC, et al. Dynamic exchange at regulatory elements during chromatin remodeling underlies assisted loading mechanism. Cell. 2011;146(4):544–54. doi: 10.1016/j.cell.2011.07.006 21835447

71. Swinstead EE, Paakinaho V, Presman DM, Hager GL. Pioneer factors and ATP-dependent chromatin remodeling factors interact dynamically: A new perspective: Multiple transcription factors can effect chromatin pioneer functions through dynamic interactions with ATP-dependent chromatin remodeling factors. Bioessays. 2016;38(11):1150–7. doi: 10.1002/bies.201600137 27633730

72. Joels M. Corticosteroid effects in the brain: U-shape it. Trends Pharmacol Sci. 2006;27(5):244–50. doi: 10.1016/j.tips.2006.03.007 16584791

73. Cairns W, Cairns C, Pongratz I, Poellinger L, Okret S. Assembly of a glucocorticoid receptor complex prior to DNA binding enhances its specific interaction with a glucocorticoid response element. J Biol Chem. 1991;266(17):11221–6. 2040629

74. Tsugita M, Iwasaki Y, Nishiyama M, Taguchi T, Shinahara M, Taniguchi Y, et al. Glucocorticoid receptor plays an indispensable role in mineralocorticoid receptor-dependent transcription in GR-deficient BE(2)C and T84 cells in vitro. Mol Cell Endocrinol. 2009;302(1):18–25. doi: 10.1016/j.mce.2008.12.008 19146914

75. van Tilborg MA, Lefstin JA, Kruiskamp M, Teuben J, Boelens R, Yamamoto KR, et al. Mutations in the glucocorticoid receptor DNA-binding domain mimic an allosteric effect of DNA. J Mol Biol. 2000;301(4):947–58. doi: 10.1006/jmbi.2000.4001 10966797

76. Watson LC, Kuchenbecker KM, Schiller BJ, Gross JD, Pufall MA, Yamamoto KR. The glucocorticoid receptor dimer interface allosterically transmits sequence-specific DNA signals. Nat Struct Mol Biol. 2013;20(7):876–83. doi: 10.1038/nsmb.2595 23728292

77. Helsen C, Claessens F. Looking at nuclear receptors from a new angle. Mol Cell Endocrinol. 2014;382(1):97–106. doi: 10.1016/j.mce.2013.09.009 24055275

78. Rogatsky I, Wang JC, Derynck MK, Nonaka DF, Khodabakhsh DB, Haqq CM, et al. Target-specific utilization of transcriptional regulatory surfaces by the glucocorticoid receptor. Proc Natl Acad Sci U S A. 2003;100(24):13845–50. doi: 10.1073/pnas.2336092100 14617768

79. Muchardt C, Yaniv M. A human homologue of Saccharomyces cerevisiae SNF2/SWI2 and Drosophila brm genes potentiates transcriptional activation by the glucocorticoid receptor. EMBO J. 1993;12(11):4279–90. 8223438

80. Hall JM, McDonnell DP, Korach KS. Allosteric regulation of estrogen receptor structure, function, and coactivator recruitment by different estrogen response elements. Mol Endocrinol. 2002;16(3):469–86. doi: 10.1210/mend.16.3.0814 11875105

81. Presman DM, Alvarez LD, Levi V, Eduardo S, Digman MA, Marti MA, et al. Insights on glucocorticoid receptor activity modulation through the binding of rigid steroids. PLoS One. 2010;5(10):e13279. doi: 10.1371/journal.pone.0013279 20949009

82. Lombes M, Binart N, Oblin ME, Joulin V, Baulieu EE. Characterization of the interaction of the human mineralocorticosteroid receptor with hormone response elements. Biochem J. 1993;292 (Pt 2):577–83.

83. Grossmann C, Ruhs S, Langenbruch L, Mildenberger S, Stratz N, Schumann K, et al. Nuclear shuttling precedes dimerization in mineralocorticoid receptor signaling. Chem Biol. 2012;19(6):742–51. doi: 10.1016/j.chembiol.2012.04.014 22726688

84. Schaaf MJ, Cidlowski JA. Molecular determinants of glucocorticoid receptor mobility in living cells: the importance of ligand affinity. Mol Cell Biol. 2003;23(6):1922–34. doi: 10.1128/MCB.23.6.1922-1934.2003 12612067

85. Chen ZP, Iyer J, Bourguet W, Held P, Mioskowski C, Lebeau L, et al. Ligand- and DNA-induced dissociation of RXR tetramers. J Mol Biol. 1998;275(1):55–65. doi: 10.1006/jmbi.1997.1413 9451439

86. Derfoul A, Robertson NM, Hall DJ, Litwack G. The N-terminal domain of the mineralocorticoid receptor modulates both mineralocorticoid receptor- and glucocorticoid receptor-mediated transactivation from Na/K ATPase beta1 target gene promoter. Endocrine. 2000;13(3):287–95. doi: 10.1385/ENDO:13:3:287 11216640

87. Rebuffat AG, Tam S, Nawrocki AR, Baker ME, Frey BM, Frey FJ, et al. The 11-ketosteroid 11-ketodexamethasone is a glucocorticoid receptor agonist. Mol Cell Endocrinol. 2004;214(1–2):27–37. doi: 10.1016/j.mce.2003.11.027 15062542

88. Medina A, Seasholtz AF, Sharma V, Burke S, Bunney W Jr., Myers RM, et al. Glucocorticoid and mineralocorticoid receptor expression in the human hippocampus in major depressive disorder. J Psychiatr Res. 2013;47(3):307–14. doi: 10.1016/j.jpsychires.2012.11.002 23219281

89. Oakley RH, Cruz-Topete D, He B, Foley JF, Myers PH, Xu X, et al. Cardiomyocyte glucocorticoid and mineralocorticoid receptors directly and antagonistically regulate heart disease in mice. Sci Signal. 2019;12(577).

90. Hoppmann J, Perwitz N, Meier B, Fasshauer M, Hadaschik D, Lehnert H, et al. The balance between gluco- and mineralo-corticoid action critically determines inflammatory adipocyte responses. J Endocrinol. 2010;204(2):153–64. doi: 10.1677/JOE-09-0292 19939912

91. Gouilleux F, Sola B, Couette B, Richard-Foy H. Cooperation between structural elements in hormono-regulated transcription from the mouse mammary tumor virus promoter. Nucleic Acids Res. 1991;19(7):1563–9. doi: 10.1093/nar/19.7.1563 1851294

92. Voss TC, Schiltz RL, Sung MH, Johnson TA, John S, Hager GL. Combinatorial probabilistic chromatin interactions produce transcriptional heterogeneity. J Cell Sci. 2009;122(Pt 3):345–56. doi: 10.1242/jcs.035865 19126674

93. Swift S, Lorens J, Achacoso P, Nolan GP. Rapid production of retroviruses for efficient gene delivery to mammalian cells using 293T cell-based systems. Curr Protoc Immunol. 2001;Chapter 10:Unit 10 7C.

94. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339(6121):819–23. doi: 10.1126/science.1231143 23287718

95. van der Valk JB, Vijverberg HP. Neuroblastoma cells as possible model in the study of glutamate receptors. Amino Acids. 1991;1(1):91–5. doi: 10.1007/BF00808095 24194051

96. Berman HM, Westbrook J, Feng Z, Gilliland G, Bhat TN, Weissig H, et al. The Protein Data Bank. Nucleic Acids Res. 2000;28(1):235–42. doi: 10.1093/nar/28.1.235 10592235

97. Clancy JW, Zhang Y, Sheehan C, D'Souza-Schorey C. An ARF6-Exportin-5 axis delivers pre-miRNA cargo to tumour microvesicles. Nat Cell Biol. 2019;21(7):856–66. doi: 10.1038/s41556-019-0345-y 31235936

98. Lubner CE, Jennings DP, Mulder DW, Schut GJ, Zadvornyy OA, Hoben JP, et al. Mechanistic insights into energy conservation by flavin-based electron bifurcation. Nat Chem Biol. 2017;13(6):655–9. doi: 10.1038/nchembio.2348 28394885

99. Pierce B, Weng Z. ZRANK: reranking protein docking predictions with an optimized energy function. Proteins. 2007;67(4):1078–86. doi: 10.1002/prot.21373 17373710

100. Krissinel E, Henrick K. Inference of macromolecular assemblies from crystalline state. J Mol Biol. 2007;372(3):774–97. doi: 10.1016/j.jmb.2007.05.022 17681537

101. Waterhouse A, Bertoni M, Bienert S, Studer G, Tauriello G, Gumienny R, et al. SWISS-MODEL: homology modelling of protein structures and complexes. Nucleic Acids Res. 2018;46(W1):W296–W303. doi: 10.1093/nar/gky427 29788355

102. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Basic local alignment search tool. J Mol Biol. 1990;215(3):403–10. doi: 10.1016/S0022-2836(05)80360-2 2231712

103. Bogan AA, Thorn KS. Anatomy of hot spots in protein interfaces. J Mol Biol. 1998;280(1):1–9. doi: 10.1006/jmbi.1998.1843 9653027

104. Cavga AD, Karahan N, Keskin O, Gursoy A. Taming Oncogenic Signaling at Protein Interfaces: Challenges and Opportunities. Curr Top Med Chem. 2015;15(20):2005–18. doi: 10.2174/1568026615666150519101956 25986691

105. Cukuroglu E, Gursoy A, Keskin O. HotRegion: a database of predicted hot spot clusters. Nucleic Acids Res. 2012;40(Database issue):D829–33. doi: 10.1093/nar/gkr929 22080558

106. Schymkowitz J, Borg J, Stricher F, Nys R, Rousseau F, Serrano L. The FoldX web server: an online force field. Nucleic Acids Res. 2005;33(Web Server issue):W382–8. doi: 10.1093/nar/gki387 15980494

107. Van Durme J, Delgado J, Stricher F, Serrano L, Schymkowitz J, Rousseau F. A graphical interface for the FoldX forcefield. Bioinformatics. 2011;27(12):1711–2. doi: 10.1093/bioinformatics/btr254 21505037

108. Dehouck Y, Kwasigroch JM, Rooman M, Gilis D. BeAtMuSiC: Prediction of changes in protein-protein binding affinity on mutations. Nucleic Acids Res. 2013;41(Web Server issue):W333–9. doi: 10.1093/nar/gkt450 23723246

109. Pires DE, Ascher DB, Blundell TL. mCSM: predicting the effects of mutations in proteins using graph-based signatures. Bioinformatics. 2014;30(3):335–42. doi: 10.1093/bioinformatics/btt691 24281696

110. Li M, Simonetti FL, Goncearenco A, Panchenko AR. MutaBind estimates and interprets the effects of sequence variants on protein-protein interactions. Nucleic Acids Res. 2016;44(W1):W494–501. doi: 10.1093/nar/gkw374 27150810


Článok vyšiel v časopise

PLOS One


2020 Číslo 1
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#