Controlling Gene Expression in Hypoxia: Comparison
Please note this is a comparison between Version 2 by Lily Guo and Version 1 by Michael Batie.

Hypoxia—reduction in oxygen availability—plays key roles in both physiological and pathological processes. Given the importance of oxygen for cell and organism viability, mechanisms to sense and respond to hypoxia are in place. A variety of enzymes utilise molecular oxygen, but of particular importance to oxygen sensing are the 2-oxoglutarate (2-OG) dependent dioxygenases (2-OGDs). Of these, Prolyl-hydroxylases have long been recognised to control the levels and function of Hypoxia Inducible Factor (HIF), a master transcriptional regulator in hypoxia, via their hydroxylase activity. However, recent studies are revealing that dioxygenases are involved in almost all aspects of gene regulation, including chromatin organisation, transcription and translation. We highlight the relevance of HIF and 2-OGDs in the control of gene expression in response to hypoxia and their relevance to human biology and health. 

  • hypoxia
  • 2-OG dioxygenases
  • HIF
  • transcription
  • trmas
  • translation
  • chromatin
Please wait, diff process is still running!

References

  1. Semenza, G.L.; Wang, G.L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythro-poietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 1992, 12, 5447–5454, doi:10.1128/mcb.12.12.5447.
  2. Rocha, S. Gene regulation under low oxygen: Holding your breath for transcription. Trends Biochem. Sci. 2007, 32, 389–397, doi:10.1016/j.tibs.2007.06.005.
  3. Duan, C. Hypoxia-inducible factor 3 biology: Complexities and emerging themes. Am. J. Physiol. Cell Physiol. 2016, 310, C260–269, doi:10.1152/ajpcell.00315.2015.
  4. Kliewe, F.; Engelhardt, M.; Aref, R.; Schuller, H.J. Promoter recruitment of corepressors Sin3 and Cyc8 by activator proteins of the yeast Saccharomyces cerevisiae. Curr. Genet. 2017, 63, 739–750, doi:10.1007/s00294-017-0677-8.
  5. D'Ignazio, L.; Batie, M.; Rocha, S. Hypoxia and Inflammation in Cancer, Focus on HIF and NF-kappaB. Biomedicines 2017, 5, 21, doi:10.3390/biomedicines5020021.
  6. Biddlestone, J.; Bandarra, D.; Rocha, S. The role of hypoxia in inflammatory disease (review). Int. J. Mol. Med. 2015, 35, 859–869, doi:10.3892/ijmm.2015.2079.
  7. Epstein, A.C.; Gleadle, J.M.; McNeill, L.A.; Hewitson, K.S.; O'Rourke, J.; Mole, D.R.; Mukherji, M.; Metzen, E.; Wilson, M.I.; Dhanda, A.; et al. C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 2001, 107, 43–54, doi:10.1016/s0092-8674(01)00507-4.
  8. Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando, J.; Ohh, M.; Salic, A.; Asara, J.M.; Lane, W.S.; Kaelin, W.G., Jr. HIFalpha targeted for VHL-mediated destruction by proline hydroxylation: Implications for O2 sensing. Science 2001, 292, 464–468, doi:10.1126/science.1059817.
  9. Maxwell, P.H.; Wiesener, M.S.; Chang, G.W.; Clifford, S.C.; Vaux, E.C.; Cockman, M.E.; Wykoff, C.C.; Pugh, C.W.; Maher, E.R.; Ratcliffe, P.J. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 1999, 399, 271–275, doi:10.1038/20459.
  10. Ohh, M.; Park, C.W.; Ivan, M.; Hoffman, M.A.; Kim, T.Y.; Huang, L.E.; Pavletich, N.; Chau, V.; Kaelin, W.G. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat. Cell Biol. 2000, 2, 423–427, doi:10.1038/35017054.
  11. Kaelin, W.G., Jr.; Ratcliffe, P.J. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol. Cell 2008, 30, 393–402, doi:10.1016/j.molcel.2008.04.009.
  12. Ehrismann, D.; Flashman, E.; Genn, D.N.; Mathioudakis, N.; Hewitson, K.S.; Ratcliffe, P.J.; Schofield, C.J. Studies on the ac-tivity of the hypoxia-inducible-factor hydroxylases using an oxygen consumption assay. Biochem. J. 2007, 401, 227–234, doi:10.1042/BJ20061151.
  13. Choudhry, H.; Harris, A.L. Advances in Hypoxia-Inducible Factor Biology. Cell Metab. 2018, 27, 281–298, doi:10.1016/j.cmet.2017.10.005.
  14. Wilson, J.W.; Shakir, D.; Batie, M.; Frost, M.; Rocha, S. Oxygen-sensing mechanisms in cells. FEBS J. 2020, 10.1111/febs.15374, doi:10.1111/febs.15374.
  15. Batie, M.; Rocha, S. Gene transcription and chromatin regulation in hypoxia. Biochem. Soc. Trans. 2020, 10.1042/BST20191106, doi:10.1042/BST20191106.
  16. Schodel, J.; Ratcliffe, P.J. Mechanisms of hypoxia signalling: New implications for nephrology. Nat. Rev. Nephrol. 2019, 15, 641–659, doi:10.1038/s41581-019-0182-z.
  17. Ortiz-Barahona, A.; Villar, D.; Pescador, N.; Amigo, J.; del Peso, L. Genome-wide identification of hypoxia-inducible factor binding sites and target genes by a probabilistic model integrating transcription-profiling data and in silico binding site prediction. Nucleic Acids Res. 2010, 38, 2332–2345, doi:10.1093/nar/gkp1205.
  18. Benita, Y.; Kikuchi, H.; Smith, A.D.; Zhang, M.Q.; Chung, D.C.; Xavier, R.J. An integrative genomics approach identifies Hypoxia Inducible Factor-1 (HIF-1)-target genes that form the core response to hypoxia. Nucleic Acids Res. 2009, 37, 4587–4602, doi:10.1093/nar/gkp425.
  19. Dengler, V.L.; Galbraith, M.; Espinosa, J.M. Transcriptional regulation by hypoxia inducible factors. Crit. Rev. Biochem. Mol. Biol. 2014, 49, 1–15, doi:10.3109/10409238.2013.838205.
  20. Schodel, J.; Oikonomopoulos, S.; Ragoussis, J.; Pugh, C.W.; Ratcliffe, P.J.; Mole, D.R. High-resolution genome-wide mapping of HIF-binding sites by ChIP-seq. Blood 2011, 117, e207–217, doi:10.1182/blood-2010-10-314427.
  21. Platt, J.L.; Salama, R.; Smythies, J.; Choudhry, H.; Davies, J.O.; Hughes, J.R.; Ratcliffe, P.J.; Mole, D.R. Capture-C reveals preformed chromatin interactions between HIF-binding sites and distant promoters. EMBO Rep. 2016, 17, 1410–1421, doi:10.15252/embr.201642198.
  22. Orlando, I.M.C.; Lafleur, V.N.; Storti, F.; Spielmann, P.; Crowther, L.; Santambrogio, S.; Schodel, J.; Hoogewijs, D.; Mole, D.R.; Wenger, R.H. Distal and proximal hypoxia response elements cooperate to regulate organ-specific erythropoietin gene expression. Haematologica 2019, doi:10.3324/haematol.2019.236406.
  23. Kenneth, N.S.; Rocha, S. Regulation of gene expression by hypoxia. Biochem. J. 2008, 414, 19–29, doi:10.1042/BJ20081055.
  24. Batie, M.; Del Peso, L.; Rocha, S. Hypoxia and Chromatin: A Focus on Transcriptional Repression Mechanisms. Biomedicines 2018, 6, 47, doi:10.3390/biomedicines6020047.
  25. Metzen, E.; Stiehl, D.P.; Doege, K.; Marxsen, J.H.; Hellwig-Burgel, T.; Jelkmann, W. Regulation of the prolyl hydroxylase domain protein 2 (phd2/egln-1) gene: Identification of a functional hypoxia-responsive element. Biochem. J. 2005, 387, 711–717, doi:10.1042/BJ20041736.
  26. Pescador, N.; Cuevas, Y.; Naranjo, S.; Alcaide, M.; Villar, D.; Landazuri, M.O.; Del Peso, L. Identification of a functional hypoxia-responsive element that regulates the expression of the egl nine homologue 3 (egln3/phd3) gene. Biochem. J. 2005, 390, 189–197, doi:10.1042/BJ20042121.
  27. Shmakova, A.; Batie, M.; Druker, J.; Rocha, S. Chromatin and oxygen sensing in the context of JmjC histone demethylases. Biochem. J. 2014, 462, 385–395, doi:10.1042/BJ20140754.
  28. Wu, M.Z.; Chen, S.F.; Nieh, S.; Benner, C.; Ger, L.P.; Jan, C.I.; Ma, L.; Chen, C.H.; Hishida, T.; Chang, H.T.; et al. Hypoxia Drives Breast Tumor Malignancy through a TET-TNFalpha-p38-MAPK Signaling Axis. Cancer Res. 2015, 75, 3912–3924, doi:10.1158/0008-5472.CAN-14-3208.
  29. Cao, J.Z.; Liu, H.; Wickrema, A.; Godley, L.A. HIF-1 directly induces TET3 expression to enhance 5-hmC density and induce erythroid gene expression in hypoxia. Blood Adv. 2020, 4, 3053–3062, doi:10.1182/bloodadvances.2020001535.
  30. Hu, R.; Jin, H.; Zhou, S.; Yang, P.; Li, X. Proteomic analysis of hypoxia-induced responses in the syncytialization of human placental cell line BeWo. Placenta 2007, 28, 399–407, doi:10.1016/j.placenta.2006.07.005.
  31. Sharma, N.K.; Sethy, N.K.; Bhargava, K. Comparative proteome analysis reveals differential regulation of glycolytic and antioxidant enzymes in cortex and hippocampus exposed to short-term hypobaric hypoxia. J. Proteom. 2013, 79, 277–298, doi:10.1016/j.jprot.2012.12.020.
  32. Hoang, V.M.; Foulk, R.; Clauser, K.; Burlingame, A.; Gibson, B.W.; Fisher, S.J. Functional proteomics: Examining the effects of hypoxia on the cytotrophoblast protein repertoire. Biochemistry 2001, 40, 4077–4086, doi:10.1021/bi0023910.
  33. Li, Q.; Luo, T.; Lu, W.; Yi, X.; Zhao, Z.; Liu, J. Proteomic analysis of human periodontal ligament cells under hypoxia. Proteome Sci. 2019, 17, 3, doi:10.1186/s12953-019-0151-2.
  34. Dou, L.; Yan, Q.; Liang, P.; Zhou, P.; Zhang, Y.; Ji, P. iTRAQ-Based Proteomic Analysis Exploring the Influence of Hypoxia on the Proteome of Dental Pulp Stem Cells under 3D Culture. Proteomics 2018, 18, doi:10.1002/pmic.201700215.
  35. Dutta, B.; Ren, Y.; Hao, P.; Sim, K.H.; Cheow, E.; Adav, S.; Tam, J.P.; Sze, S.K. Profiling of the Chromatin-associated Proteome Identifies HP1BP3 as a Novel Regulator of Cell Cycle Progression. Mol. Cell. Proteom. 2014, 13, 2183–2197, doi:10.1074/mcp.M113.034975.
  36. Gao, Y.; Dasgupta, C.; Huang, L.; Song, R.; Zhang, Z.; Zhang, L. Multi-Omics Integration Reveals Short and Long-Term Effects of Gestational Hypoxia on the Heart Development. Cells 2019, 8, 608, doi:10.3390/cells8121608.
  37. Shakib, K.; Norman, J.T.; Fine, L.G.; Brown, L.R.; Godovac-Zimmermann, J. Proteomics profiling of nuclear proteins for kidney fibroblasts suggests hypoxia, meiosis, and cancer may meet in the nucleus. Proteomics 2005, 5, 2819–2838, doi:10.1002/pmic.200401108.
  38. Vodisch, M.; Scherlach, K.; Winkler, R.; Hertweck, C.; Braun, H.P.; Roth, M.; Haas, H.; Werner, E.R.; Brakhage, A.A.; Kniemeyer, O. Analysis of the Aspergillus fumigatus proteome reveals metabolic changes and the activation of the pseurotin A biosynthesis gene cluster in response to hypoxia. J. Proteome Res. 2011, 10, 2508–2524, doi:10.1021/pr1012812.
  39. Janker, L.; Mayer, R.L.; Bileck, A.; Kreutz, D.; Mader, J.C.; Utpatel, K.; Heudobler, D.; Agis, H.; Gerner, C.; Slany, A. Metabolic, Anti-apoptotic and Immune Evasion Strategies of Primary Human Myeloma Cells Indicate Adaptations to Hypoxia. Mol. Cell. Proteom. 2019, 18, 936–953, doi:10.1074/mcp.RA119.001390.
  40. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705, doi:10.1016/j.cell.2007.02.005.
  41. Hsu, K.F.; Wilkins, S.E.; Hopkinson, R.J.; Sekirnik, R.; Flashman, E.; Kawamura, A.; McCullagh, J.S.O.; Walport, L.J.; Schofield, C.J. Hypoxia and hypoxia mimetics differentially modulate histone post-translational modifications. Epigenetics 2020, doi:10.1080/15592294.2020.1786305.
  42. Batie, M.; Frost, J.; Frost, M.; Wilson, J.W.; Schofield, P.; Rocha, S. Hypoxia induces rapid changes to histone methylation and reprograms chromatin. Science 2019, 363, 1222–1226, doi:10.1126/science.aau5870.
  43. Prickaerts, P.; Adriaens, M.E.; Beucken, T.V.; Koch, E.; Dubois, L.; Dahlmans, V.E.; Gits, C.; Evelo, C.T.; Chan-Seng-Yue, M.; Wouters, B.G.; et al. Hypoxia increases genome-wide bivalent epigenetic marking by specific gain of H3K27me3. Epigenet. Chromatin 2016, 9, 46, doi:10.1186/s13072-016-0086-0.
  44. Chakraborty, A.A.; Laukka, T.; Myllykoski, M.; Ringel, A.E.; Booker, M.A.; Tolstorukov, M.Y.; Meng, Y.J.; Meier, S.R.; Jen-nings, R.B.; Creech, A.L.; et al. Histone demethylase KDM6A directly senses oxygen to control chromatin and cell fate. Sci-ence 2019, 363, 1217–1222, doi:10.1126/science.aaw1026.
  45. Lee, H.Y.; Yang, E.G.; Park, H. Hypoxia enhances the expression of prostate-specific antigen by modifying the quantity and catalytic activity of Jumonji C domain-containing histone demethylases. Carcinogenesis 2013, 34, 2706–2715, doi:10.1093/carcin/bgt256.
  46. Dobrynin, G.; McAllister, T.E.; Leszczynska, K.B.; Ramachandran, S.; Krieg, A.J.; Kawamura, A.; Hammond, E.M. KDM4A regulates HIF-1 levels through H3K9me3. Sci. Rep. 2017, 7, 11094, doi:10.1038/s41598-017-11658-3.
  47. Luo, W.; Chang, R.; Zhong, J.; Pandey, A.; Semenza, G.L. Histone demethylase JMJD2C is a coactivator for hypoxia-inducible factor 1 that is required for breast cancer progression. Proc. Natl. Acad. Sci. USA 2012, 109, E3367-3376, doi:10.1073/pnas.1217394109.
  48. Krieg, A.J.; Rankin, E.B.; Chan, D.; Razorenova, O.; Fernandez, S.; Giaccia, A.J. Regulation of the histone demethylase JMJD1A by hypoxia-inducible factor 1 alpha enhances hypoxic gene expression and tumor growth. Mol. Cell. Biol. 2010, 30, 344–353, doi:10.1128/MCB.00444-09.
  49. Wan, W.; Peng, K.; Li, M.; Qin, L.; Tong, Z.; Yan, J.; Shen, B.; Yu, C. Histone demethylase JMJD1A promotes urinary bladder cancer progression by enhancing glycolysis through coactivation of hypoxia inducible factor 1alpha. Oncogene 2017, 36, 3868–3877, doi:10.1038/onc.2017.13.
  50. Qian, X.; Li, X.; Shi, Z.; Bai, X.; Xia, Y.; Zheng, Y.; Xu, D.; Chen, F.; You, Y.; Fang, J.; et al. KDM3A Senses Oxygen Availability to Regulate PGC-1alpha-Mediated Mitochondrial Biogenesis. Mol. Cell. 2019, 76, 885–895 e887, doi:10.1016/j.molcel.2019.09.019.
  51. Wu, X.; Zhang, Y. TET-mediated active DNA demethylation: Mechanism, function and beyond. Nat. Rev. Genet. 2017, 18, 517–534, doi:10.1038/nrg.2017.33.
  52. Thienpont, B.; Steinbacher, J.; Zhao, H.; D'Anna, F.; Kuchnio, A.; Ploumakis, A.; Ghesquiere, B.; Van Dyck, L.; Boeckx, B.; Schoonjans, L.; et al. Tumour hypoxia causes DNA hypermethylation by reducing TET activity. Nature 2016, 537, 63–68, doi:10.1038/nature19081.
  53. Shahrzad, S.; Bertrand, K.; Minhas, K.; Coomber, B.L. Induction of DNA hypomethylation by tumor hypoxia. Epigenetics 2007, 2, 119–125, doi:10.4161/epi.2.2.4613.
  54. D'Anna, F.; Van Dyck, L.; Xiong, J.; Zhao, H.; Berrens, R.V.; Qian, J.; Bieniasz-Krzywiec, P.; Chandra, V.; Schoonjans, L.; Matthews, J.; et al. DNA methylation repels binding of hypoxia-inducible transcription factors to maintain tumor immunotolerance. Genome Biol. 2020, 21, 182, doi:10.1186/s13059-020-02087-z.
  55. Ito, S.; D'Alessio, A.C.; Taranova, O.V.; Hong, K.; Sowers, L.C.; Zhang, Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 2010, 466, 1129–1133, doi:10.1038/nature09303.
  56. Camuzi, D.; de Amorim, I.S.S.; Ribeiro Pinto, L.F.; Oliveira Trivilin, L.; Mencalha, A.L.; Soares Lima, S.C. Regulation Is in the Air: The Relationship between Hypoxia and Epigenetics in Cancer. Cells 2019, 8, 300, doi:10.3390/cells8040300.
  57. Tahiliani, M.; Koh, K.P.; Shen, Y.; Pastor, W.A.; Bandukwala, H.; Brudno, Y.; Agarwal, S.; Iyer, L.M.; Liu, D.R.; Aravind, L.; et al. Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science 2009, 324, 930–935, doi:10.1126/science.1170116.
  58. Brugarolas, J.; Lei, K.; Hurley, R.L.; Manning, B.D.; Reiling, J.H.; Hafen, E.; Witters, L.A.; Ellisen, L.W.; Kaelin, W.G., Jr. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex. Genes Dev. 2004, 18, 2893–2904, doi:10.1101/gad.1256804.
  59. Connolly, E.; Braunstein, S.; Formenti, S.; Schneider, R.J. Hypoxia inhibits protein synthesis through a 4E-BP1 and elongation factor 2 kinase pathway controlled by mTOR and uncoupled in breast cancer cells. Mol. Cell. Biol. 2006, 26, 3955–3965, doi:10.1128/MCB.26.10.3955-3965.2006.
  60. Sonenberg, N.; Hinnebusch, A.G. Regulation of translation initiation in eukaryotes: Mechanisms and biological targets. Cell 2009, 136, 731–745, doi:10.1016/j.cell.2009.01.042.
  61. Koumenis, C.; Naczki, C.; Koritzinsky, M.; Rastani, S.; Diehl, A.; Sonenberg, N.; Koromilas, A.; Wouters, B.G. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2alpha. Mol. Cell. Biol. 2002, 22, 7405–7416, doi:10.1128/mcb.22.21.7405-7416.2002.
  62. Noma, A.; Ishitani, R.; Kato, M.; Nagao, A.; Nureki, O.; Suzuki, T. Expanding role of the jumonji C domain as an RNA hydroxylase. J. Biol. Chem. 2010, 285, 34503–34507, doi:10.1074/jbc.M110.156398.
  63. Donnelly, N.; Gorman, A.M.; Gupta, S.; Samali, A. The eIF2alpha kinases: Their structures and functions. Cell. Mol. Life Sci.2013, 70, 3493-3511, doi:10.1007/s00018-012-1252-6.
  64. Ryazanov, A.G.; Davydova, E.K. Mechanism of elongation factor 2 (EF-2) inactivation upon phosphorylation. Phosphorylated EF-2 is unable to catalyze translocation. FEBS Lett. 1989, 251, 187–190, doi:10.1016/0014-5793(89)81452-8.
  65. Browne, G.J.; Proud, C.G. A novel mTOR-regulated phosphorylation site in elongation factor 2 kinase modulates the activity of the kinase and its binding to calmodulin. Mol. Cell. Biol. 2004, 24, 2986–2997, doi:10.1128/mcb.24.7.2986-2997.2004.
  66. Moore, C.E.; Mikolajek, H.; Regufe da Mota, S.; Wang, X.; Kenney, J.W.; Werner, J.M.; Proud, C.G. Elongation Factor 2 Kinase Is Regulated by Proline Hydroxylation and Protects Cells during Hypoxia. Mol. Cell. Biol. 2015, 35, 1788–1804, doi:10.1128/MCB.01457-14.
  67. Pollard, P.J.; Loenarz, C.; Mole, D.R.; McDonough, M.A.; Gleadle, J.M.; Schofield, C.J.; Ratcliffe, P.J. Regulation of Jumonji-domain-containing histone demethylases by hypoxia-inducible factor (HIF)-1alpha. Biochem. J. 2008, 416, 387–394, doi:10.1042/BJ20081238.
  68. Kato, M.; Araiso, Y.; Noma, A.; Nagao, A.; Suzuki, T.; Ishitani, R.; Nureki, O. Crystal structure of a novel JmjC-domain-containing protein, TYW5, involved in tRNA modification. Nucleic Acids Res. 2011, 39, 1576–1585, doi:10.1093/nar/gkq919.
  69. Ge, W.; Wolf, A.; Feng, T.; Ho, C.H.; Sekirnik, R.; Zayer, A.; Granatino, N.; Cockman, M.E.; Loenarz, C.; Loik, N.D.; et al. Oxygenase-catalyzed ribosome hydroxylation occurs in prokaryotes and humans. Nat. Chem. Biol. 2012, 8, 960–962, doi:10.1038/nchembio.1093.
  70. Loenarz, C.; Sekirnik, R.; Thalhammer, A.; Ge, W.; Spivakovsky, E.; Mackeen, M.M.; McDonough, M.A.; Cockman, M.E.; Kessler, B.M.; Ratcliffe, P.J.; et al. Hydroxylation of the eukaryotic ribosomal decoding center affects translational accuracy. Proc. Natl. Acad. Sci. USA 2014, 111, 4019–4024, doi:10.1073/pnas.1311750111.
  71. Singleton, R.S.; Liu-Yi, P.; Formenti, F.; Ge, W.; Sekirnik, R.; Fischer, R.; Adam, J.; Pollard, P.J.; Wolf, A.; Thalhammer, A.; et al. OGFOD1 catalyzes prolyl hydroxylation of RPS23 and is involved in translation control and stress granule formation. Proc. Natl. Acad. Sci. USA 2014, 111, 4031–4036, doi:10.1073/pnas.1314482111.
  72. Fu, D.; Brophy, J.A.; Chan, C.T.; Atmore, K.A.; Begley, U.; Paules, R.S.; Dedon, P.C.; Begley, T.J.; Samson, L.D. Human AlkB homolog ABH8 Is a tRNA methyltransferase required for wobble uridine modification and DNA damage survival. Mol. Cell. Biol. 2010, 30, 2449–2459, doi:10.1128/MCB.01604-09.
  73. Liu, L.; Cash, T.P.; Jones, R.G.; Keith, B.; Thompson, C.B.; Simon, M.C. Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol. Cell. 2006, 21, 521–531, doi:10.1016/j.molcel.2006.01.010.
  74. Feng, T.; Yamamoto, A.; Wilkins, S.E.; Sokolova, E.; Yates, L.A.; Munzel, M.; Singh, P.; Hopkinson, R.J.; Fischer, R.; Cockman, M.E.; et al. Optimal translational termination requires C4 lysyl hydroxylation of eRF1. Mol. Cell. 2014, 53, 645–654, doi:10.1016/j.molcel.2013.12.028.
  75. Chang, B.; Chen, Y.; Zhao, Y.; Bruick, R.K. JMJD6 is a histone arginine demethylase. Science 2007, 318, 444–447, doi:10.1126/science.1145801.
  76. Islam, M.S.; McDonough, M.A.; Chowdhury, R.; Gault, J.; Khan, A.; Pires, E.; Schofield, C.J. Biochemical and structural investigations clarify the substrate selectivity of the 2-oxoglutarate oxygenase JMJD6. J. Biol. Chem. 2019, 294, 11637–11652, doi:10.1074/jbc.RA119.008693.
  77. Alahari, S.; Post, M.; Caniggia, I. Jumonji Domain Containing Protein 6: A Novel Oxygen Sensor in the Human Placenta. Endocrinology 2015, 156, 3012–3025, doi:10.1210/en.2015-1262.
  78. Tsai, W.C.; Reineke, L.C.; Jain, A.; Jung, S.Y.; Lloyd, R.E. Histone arginine demethylase JMJD6 is linked to stress granule assembly through demethylation of the stress granule-nucleating protein G3BP1. J. Biol. Chem. 2017, 292, 18886–18896, doi:10.1074/jbc.M117.800706.
  79. Tsai, W.C.; Gayatri, S.; Reineke, L.C.; Sbardella, G.; Bedford, M.T.; Lloyd, R.E. Arginine Demethylation of G3BP1 Promotes Stress Granule Assembly. J. Biol. Chem. 2016, 291, 22671–22685, doi:10.1074/jbc.M116.739573.
  80. Webby, C.J.; Wolf, A.; Gromak, N.; Dreger, M.; Kramer, H.; Kessler, B.; Nielsen, M.L.; Schmitz, C.; Butler, D.S.; Yates, J.R., 3rd; et al. Jmjd6 catalyses lysyl-hydroxylation of U2AF65, a protein associated with RNA splicing. Science 2009, 325, 90–93, doi:10.1126/science.1175865.
  81. Kwok, J.; O'Shea, M.; Hume, D.A.; Lengeling, A. Jmjd6, a JmjC Dioxygenase with Many Interaction Partners and Pleiotropic Functions. Front. Genet. 2017, 8, 32, doi:10.3389/fgene.2017.00032.
  82. Liu, W.; Ma, Q.; Wong, K.; Li, W.; Ohgi, K.; Zhang, J.; Aggarwal, A.; Rosenfeld, M.G. Brd4 and JMJD6-associated anti-pause enhancers in regulation of transcriptional pause release. Cell 2013, 155, 1581–1595, doi:10.1016/j.cell.2013.10.056.
  83. Wang, F.; He, L.; Huangyang, P.; Liang, J.; Si, W.; Yan, R.; Han, X.; Liu, S.; Gui, B.; Li, W.; et al. JMJD6 promotes colon carcinogenesis through negative regulation of p53 by hydroxylation. PLoS Biol. 2014, 12, e1001819, doi:10.1371/journal.pbio.1001819.
  84. Han, G.; Li, J.; Wang, Y.; Li, X.; Mao, H.; Liu, Y.; Chen, C.D. The hydroxylation activity of Jmjd6 is required for its homo-oligomerization. J. Cell. Biochem. 2012, 113, 1663–1670, doi:10.1002/jcb.24035.
  85. Tibrewal, N.; Liu, T.; Li, H.; Birge, R.B. Characterization of the biochemical and biophysical properties of the phosphatidylserine receptor (PS-R) gene product. Mol. Cell. Biochem. 2007, 304, 119–125, doi:10.1007/s11010-007-9492-8.
  86. Wilkins, S.E.; Islam, M.S.; Gannon, J.M.; Markolovic, S.; Hopkinson, R.J.; Ge, W.; Schofield, C.J.; Chowdhury, R. JMJD5 is a human arginyl C-3 hydroxylase. Nat. Commun. 2018, 9, 1180, doi:10.1038/s41467-018-03410-w.
  87. Shen, J.; Xiang, X.; Chen, L.; Wang, H.; Wu, L.; Sun, Y.; Ma, L.; Gu, X.; Liu, H.; Wang, L.; et al. JMJD5 cleaves monomethylated histone H3 N-tail under DNA damaging stress. EMBO Rep. 2017, 18, 2131–2143, doi:10.15252/embr.201743892.
  88. Liu, H.; Wang, C.; Lee, S.; Deng, Y.; Wither, M.; Oh, S.; Ning, F.; Dege, C.; Zhang, Q.; Liu, X.; et al. Clipping of arginine-methylated histone tails by JMJD5 and JMJD7. Proc. Natl. Acad. Sci. USA 2017, 114, E7717-E7726, doi:10.1073/pnas.1706831114.
  89. Liu, H.; Wang, C.; Lee, S.; Ning, F.; Wang, Y.; Zhang, Q.; Chen, Z.; Zang, J.; Nix, J.; Dai, S.; et al. Specific Recognition of Arginine Methylated Histone Tails by JMJD5 and JMJD7. Sci. Rep. 2018, 8, 3275, doi:10.1038/s41598-018-21432-8.
  90. Hsia, D.A.; Tepper, C.G.; Pochampalli, M.R.; Hsia, E.Y.; Izumiya, C.; Huerta, S.B.; Wright, M.E.; Chen, H.W.; Kung, H.J.; Izumiya, Y. KDM8, a H3K36me2 histone demethylase that acts in the cyclin A1 coding region to regulate cancer cell proliferation. Proc. Natl. Acad. Sci. USA 2010, 107, 9671–9676, doi:10.1073/pnas.1000401107.
  91. Marcon, E.; Ni, Z.; Pu, S.; Turinsky, A.L.; Trimble, S.S.; Olsen, J.B.; Silverman-Gavrila, R.; Silverman-Gavrila, L.; Phanse, S.; Guo, H.; et al. Human-chromatin-related protein interactions identify a demethylase complex required for chromosome segregation. Cell Rep. 2014, 8, 297–310, doi:10.1016/j.celrep.2014.05.050.
  92. Huang, X.; Zhang, S.; Qi, H.; Wang, Z.; Chen, H.W.; Shao, J.; Shen, J. JMJD5 interacts with p53 and negatively regulates p53 function in control of cell cycle and proliferation. Biochim. Biophys. Acta 2015, 1853, 2286–2295, doi:10.1016/j.bbamcr.2015.05.026.
  93. Ishimura, A.; Terashima, M.; Tange, S.; Suzuki, T. Jmjd5 functions as a regulator of p53 signaling during mouse embryogenesis. Cell Tissue Res. 2016, 363, 723–733, doi:10.1007/s00441-015-2276-7.
  94. Markolovic, S.; Zhuang, Q.; Wilkins, S.E.; Eaton, C.D.; Abboud, M.I.; Katz, M.J.; McNeil, H.E.; Lesniak, R.K.; Hall, C.; Struwe, W.B.; et al. The Jumonji-C oxygenase JMJD7 catalyzes (3S)-lysyl hydroxylation of TRAFAC GTPases. Nat. Chem. Biol. 2018, 14, 688–695, doi:10.1038/s41589-018-0071-y.
  95. Lu, T.; Jackson, M.W.; Wang, B.; Yang, M.; Chance, M.R.; Miyagi, M.; Gudkov, A.V.; Stark, G.R. Regulation of NF-kappaB by NSD1/FBXL11-dependent reversible lysine methylation of p65. Proc. Natl. Acad. Sci. USA 2010, 107, 46–51, doi:10.1073/pnas.0912493107.
  96. Ramadoss, S.; Guo, G.; Wang, C.Y. Lysine demethylase KDM3A regulates breast cancer cell invasion and apoptosis by targeting histone and the non-histone protein p53. Oncogene 2017, 36, 47–59, doi:10.1038/onc.2016.174.
  97. Ponnaluri, V.K.; Vavilala, D.T.; Putty, S.; Gutheil, W.G.; Mukherji, M. Identification of non-histone substrates for JMJD2A-C histone demethylases. Biochem. Biophys. Res. Commun. 2009, 390, 280–284, doi:10.1016/j.bbrc.2009.09.107.
  98. Walport, L.J.; Hopkinson, R.J.; Chowdhury, R.; Schiller, R.; Ge, W.; Kawamura, A.; Schofield, C.J. Arginine demethylation is catalysed by a subset of JmjC histone lysine demethylases. Nat. Commun. 2016, 7, 11974, doi:10.1038/ncomms11974.
  99. Meyer, K.D.; Patil, D.P.; Zhou, J.; Zinoviev, A.; Skabkin, M.A.; Elemento, O.; Pestova, T.V.; Qian, S.B.; Jaffrey, S.R. 5' UTR m(6)A Promotes Cap-Independent Translation. Cell 2015, 163, 999–1010, doi:10.1016/j.cell.2015.10.012.
  100. Stenson, P.D.; Mort, M.; Ball, E.V.; Shaw, K.; Phillips, A.; Cooper, D.N. The Human Gene Mutation Database: Building a comprehensive mutation repository for clinical and molecular genetics, diagnostic testing and personalized genomic medicine. Hum. Genet. 2014, 133, 1–9, doi:10.1007/s00439-013-1358-4.
  101. Ang, S.O.; Chen, H.; Hirota, K.; Gordeuk, V.R.; Jelinek, J.; Guan, Y.; Liu, E.; Sergueeva, A.I.; Miasnikova, G.Y.; Mole, D.; et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nat. Genet. 2002, 32, 614–621, doi:10.1038/ng1019.
  102. Gordeuk, V.R.; Sergueeva, A.I.; Miasnikova, G.Y.; Okhotin, D.; Voloshin, Y.; Choyke, P.L.; Butman, J.A.; Jedlickova, K.; Prchal, J.T.; Polyakova, L.A. Congenital disorder of oxygen sensing: Association of the homozygous Chuvash polycythemia VHL mutation with thrombosis and vascular abnormalities but not tumors. Blood 2004, 103, 3924–3932, doi:10.1182/blood-2003-07-2535.
  103. Smith, T.G.; Brooks, J.T.; Balanos, G.M.; Lappin, T.R.; Layton, D.M.; Leedham, D.L.; Liu, C.; Maxwell, P.H.; McMullin, M.F.; McNamara, C.J.; et al. Mutation of von Hippel-Lindau tumour suppressor and human cardiopulmonary physiology. PLoS Med. 2006, 3, e290, doi:10.1371/journal.pmed.0030290.
  104. Rasmussen, K.D.; Helin, K. Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 2016, 30, 733–750, doi:10.1101/gad.276568.115.
  105. Delhommeau, F.; Dupont, S.; Della Valle, V.; James, C.; Trannoy, S.; Masse, A.; Kosmider, O.; Le Couedic, J.P.; Robert, F.; Alberdi, A.; et al. Mutation in TET2 in myeloid cancers. N. Engl. J. Med. 2009, 360, 2289–2301, doi:10.1056/NEJMoa0810069.
  106. Banka, S.; Lederer, D.; Benoit, V.; Jenkins, E.; Howard, E.; Bunstone, S.; Kerr, B.; McKee, S.; Lloyd, I.C.; Shears, D.; et al. Novel KDM6A (UTX) mutations and a clinical and molecular review of the X-linked Kabuki syndrome (KS2). Clin. Genet. 2015, 87, 252–258, doi:10.1111/cge.12363.
  107. Van Laarhoven, P.M.; Neitzel, L.R.; Quintana, A.M.; Geiger, E.A.; Zackai, E.H.; Clouthier, D.E.; Artinger, K.B.; Ming, J.E.; Shaikh, T.H. Kabuki syndrome genes KMT2D and KDM6A: Functional analyses demonstrate critical roles in craniofacial, heart and brain development. Hum. Mol. Genet. 2015, 24, 4443–4453, doi:10.1093/hmg/ddv180.
  108. Lederer, D.; Grisart, B.; Digilio, M.C.; Benoit, V.; Crespin, M.; Ghariani, S.C.; Maystadt, I.; Dallapiccola, B.; Verellen-Dumoulin, C. Deletion of KDM6A, a histone demethylase interacting with MLL2, in three patients with Kabuki syndrome. Am. J. Hum. Genet. 2012, 90, 119–124, doi:10.1016/j.ajhg.2011.11.021.
  109. Yang, P.; Tan, H.; Xia, Y.; Yu, Q.; Wei, X.; Guo, R.; Peng, Y.; Chen, C.; Li, H.; Mei, L.; et al. De novo exonic deletion of KDM6A in a Chinese girl with Kabuki syndrome: A case report and brief literature review. Am. J. Med. Genet. A 2016, 170, 1613–1621, doi:10.1002/ajmg.a.37634.
  110. Lindgren, A.M.; Hoyos, T.; Talkowski, M.E.; Hanscom, C.; Blumenthal, I.; Chiang, C.; Ernst, C.; Pereira, S.; Ordulu, Z.; Clericuzio, C.; et al. Haploinsufficiency of KDM6A is associated with severe psychomotor retardation, global growth restriction, seizures and cleft palate. Hum. Genet. 2013, 132, 537–552, doi:10.1007/s00439-013-1263-x.
  111. Dunwoodie, S.L. The role of hypoxia in development of the Mammalian embryo. Dev Cell. 2009, 17, 755–773, doi:10.1016/j.devcel.2009.11.008.
  112. Huang, X.; Zhang, S.; Qi, H.; Wang, Z.; Chen, H.W.; Shao, J.; Shen, J. JMJD5 interacts with p53 and negatively regulates p53 function in control of cell cycle and proliferation. Biochim. Biophys. Acta 2015, 1853, 2286–2295, doi:10.1016/j.bbamcr.2015.05.026.
  113. Ishimura, A.; Terashima, M.; Tange, S.; Suzuki, T. Jmjd5 functions as a regulator of p53 signaling during mouse embryogenesis. Cell Tissue Res. 2016, 363, 723–733, doi:10.1007/s00441-015-2276-7.
  114. Markolovic, S.; Zhuang, Q.; Wilkins, S.E.; Eaton, C.D.; Abboud, M.I.; Katz, M.J.; McNeil, H.E.; Lesniak, R.K.; Hall, C.; Struwe, W.B.; et al. The Jumonji-C oxygenase JMJD7 catalyzes (3S)-lysyl hydroxylation of TRAFAC GTPases. Nat. Chem. Biol. 2018, 14, 688–695, doi:10.1038/s41589-018-0071-y.
  115. Lu, T.; Jackson, M.W.; Wang, B.; Yang, M.; Chance, M.R.; Miyagi, M.; Gudkov, A.V.; Stark, G.R. Regulation of NF-kappaB by NSD1/FBXL11-dependent reversible lysine methylation of p65. Proc. Natl. Acad. Sci. USA 2010, 107, 46–51, doi:10.1073/pnas.0912493107.
  116. Ramadoss, S.; Guo, G.; Wang, C.Y. Lysine demethylase KDM3A regulates breast cancer cell invasion and apoptosis by targeting histone and the non-histone protein p53. Oncogene 2017, 36, 47–59, doi:10.1038/onc.2016.174.
  117. Ponnaluri, V.K.; Vavilala, D.T.; Putty, S.; Gutheil, W.G.; Mukherji, M. Identification of non-histone substrates for JMJD2A-C histone demethylases. Biochem. Biophys. Res. Commun. 2009, 390, 280–284, doi:10.1016/j.bbrc.2009.09.107.
  118. Walport, L.J.; Hopkinson, R.J.; Chowdhury, R.; Schiller, R.; Ge, W.; Kawamura, A.; Schofield, C.J. Arginine demethylation is catalysed by a subset of JmjC histone lysine demethylases. Nat. Commun. 2016, 7, 11974, doi:10.1038/ncomms11974.
  119. Meyer, K.D.; Patil, D.P.; Zhou, J.; Zinoviev, A.; Skabkin, M.A.; Elemento, O.; Pestova, T.V.; Qian, S.B.; Jaffrey, S.R. 5' UTR m(6)A Promotes Cap-Independent Translation. Cell 2015, 163, 999–1010, doi:10.1016/j.cell.2015.10.012.
  120. Stenson, P.D.; Mort, M.; Ball, E.V.; Shaw, K.; Phillips, A.; Cooper, D.N. The Human Gene Mutation Database: Building a comprehensive mutation repository for clinical and molecular genetics, diagnostic testing and personalized genomic medicine. Hum. Genet. 2014, 133, 1–9, doi:10.1007/s00439-013-1358-4.
  121. Ang, S.O.; Chen, H.; Hirota, K.; Gordeuk, V.R.; Jelinek, J.; Guan, Y.; Liu, E.; Sergueeva, A.I.; Miasnikova, G.Y.; Mole, D.; et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nat. Genet. 2002, 32, 614–621, doi:10.1038/ng1019.
  122. Gordeuk, V.R.; Sergueeva, A.I.; Miasnikova, G.Y.; Okhotin, D.; Voloshin, Y.; Choyke, P.L.; Butman, J.A.; Jedlickova, K.; Prchal, J.T.; Polyakova, L.A. Congenital disorder of oxygen sensing: Association of the homozygous Chuvash polycythemia VHL mutation with thrombosis and vascular abnormalities but not tumors. Blood 2004, 103, 3924–3932, doi:10.1182/blood-2003-07-2535.
  123. Smith, T.G.; Brooks, J.T.; Balanos, G.M.; Lappin, T.R.; Layton, D.M.; Leedham, D.L.; Liu, C.; Maxwell, P.H.; McMullin, M.F.; McNamara, C.J.; et al. Mutation of von Hippel-Lindau tumour suppressor and human cardiopulmonary physiology. PLoS Med. 2006, 3, e290, doi:10.1371/journal.pmed.0030290.
  124. Rasmussen, K.D.; Helin, K. Role of TET enzymes in DNA methylation, development, and cancer. Genes Dev. 2016, 30, 733–750, doi:10.1101/gad.276568.115.
  125. Delhommeau, F.; Dupont, S.; Della Valle, V.; James, C.; Trannoy, S.; Masse, A.; Kosmider, O.; Le Couedic, J.P.; Robert, F.; Alberdi, A.; et al. Mutation in TET2 in myeloid cancers. N. Engl. J. Med. 2009, 360, 2289–2301, doi:10.1056/NEJMoa0810069.
  126. Lederer, D.; Grisart, B.; Digilio, M.C.; Benoit, V.; Crespin, M.; Ghariani, S.C.; Maystadt, I.; Dallapiccola, B.; Verellen-Dumoulin, C. Deletion of KDM6A, a histone demethylase interacting with MLL2, in three patients with Kabuki syndrome. Am. J. Hum. Genet. 2012, 90, 119–124, doi:10.1016/j.ajhg.2011.11.021.
  127. Yang, P.; Tan, H.; Xia, Y.; Yu, Q.; Wei, X.; Guo, R.; Peng, Y.; Chen, C.; Li, H.; Mei, L.; et al. De novo exonic deletion of KDM6A in a Chinese girl with Kabuki syndrome: A case report and brief literature review. Am. J. Med. Genet. A 2016, 170, 1613–1621, doi:10.1002/ajmg.a.37634.
  128. Lindgren, A.M.; Hoyos, T.; Talkowski, M.E.; Hanscom, C.; Blumenthal, I.; Chiang, C.; Ernst, C.; Pereira, S.; Ordulu, Z.; Clericuzio, C.; et al. Haploinsufficiency of KDM6A is associated with severe psychomotor retardation, global growth restriction, seizures and cleft palate. Hum. Genet. 2013, 132, 537–552, doi:10.1007/s00439-013-1263-x.
  129. Dunwoodie, S.L. The role of hypoxia in development of the Mammalian embryo. Dev Cell. 2009, 17, 755–773, doi:10.1016/j.devcel.2009.11.008.
  130. Banka, S.; Lederer, D.; Benoit, V.; Jenkins, E.; Howard, E.; Bunstone, S.; Kerr, B.; McKee, S.; Lloyd, I.C.; Shears, D.; et al. Novel KDM6A (UTX) mutations and a clinical and molecular review of the X-linked Kabuki syndrome (KS2). Clin. Genet. 2015, 87, 252–258, doi:10.1111/cge.12363.
  131. Van Laarhoven, P.M.; Neitzel, L.R.; Quintana, A.M.; Geiger, E.A.; Zackai, E.H.; Clouthier, D.E.; Artinger, K.B.; Ming, J.E.; Shaikh, T.H. Kabuki syndrome genes KMT2D and KDM6A: Functional analyses demonstrate critical roles in craniofacial, heart and brain development. Hum. Mol. Genet. 2015, 24, 4443–4453, doi:10.1093/hmg/ddv180.
More
Video Production Service