Skip to main content

Advertisement

Log in

Hypoxia Inducible Factor Prolyl 4-Hydroxylase Enzymes: Center Stage in the Battle Against Hypoxia, Metabolic Compromise and Oxidative Stress

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

Studies of adaptive mechanisms to hypoxia led to the discovery of the transcription factor called hypoxia inducible factor (HIF). HIF is a ubiquitously expressed, heterodimeric transcription factor that regulates a cassette of genes that can provide compensation for hypoxia, metabolic compromise, and oxidative stress including erythropoietin, vascular endothelial growth factor, or glycolytic enzymes. Diseases associated with oxygen deprivation and consequent metabolic compromise such as stroke or Alzheimer’s disease may result from inadequate engagement of adaptive signaling pathways that culminate in HIF activation. The discovery that HIF stability and activation are governed by a family of dioxygenases called HIF prolyl 4 hydroxylases (PHDs) identified a new target to augment the transcriptional activity of HIF and thus the adaptive machinery that governs neuroprotection. PHDs lose activity when cells are deprived of oxygen, iron or 2-oxoglutarate. Inhibition of PHD activity triggers the cellular homeostatic response to oxygen and glucose deprivation by stabilizing HIF and other proteins. Herein, we discuss the possible role of PHDs in regulation of both HIF-dependent and -independent cell survival pathways in the nervous system with particular attention to the co-substrate requirements for these enzymes. The emergence of neuroprotective therapies that modulate genes capable of combating metabolic compromise is an affirmation of elegant studies done by John Blass and colleagues over the past five decades implicating altered metabolism in neurodegeneration.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4

Similar content being viewed by others

Abbreviations

AP-1:

Activator protein 1

CO2 :

Carbon dioxide

CoCl2 :

Cobalt chloride

CODD:

Carboxy-terminal oxygen degradation domain

DFO:

Desferrioxamine

DMOG:

Dimethyl-oxalyl-glycine

EGR-1:

Early growth response protein-1

ERK:

Extracellular signal-regulated kinase

Fe2+ :

Ferrous

Fe3+ :

Ferric

FH:

Fumarate hydratase

HIF:

Hypoxia inducible factor

HIF-1α:

Hypoxia inducible factor-1 alpha

HIF-1β:

Hypoxia inducible factor-1 beta

HRE:

Hypoxia response elements

IRP2:

Iron regulatory protein-2

LIP:

Labile iron pool

MnSOD:

Manganese Superoxide Dismutase

MORG-1:

Mitogen-activated protein kinase organizer-1

NF-kB:

Nuclear factor kappa B

NODD:

N-terminal oxygen degradation domain

O2 :

Oxygen

ODD:

Oxygen degradation domain

ODDD:

Oxygen dependent degradation domain

PHD:

prolyl 4-hydroxylase domain

PVHL:

von Hippel-Lindau protein

RNA pol II:

RNA polymerase II

ROS:

Reactive Oxygen Species

SIAH2:

Seven in absentia homolog 2

SDH:

Succinate dehydrogenase

TCA:

Tricarboxylic acid

VEGF:

Vascular endothelial growth factor

References

  1. Ikeda E (2005). Cellular response to tissue hypoxia and its involvement in disease progression. Pathol Int 55:603–610

    Article  PubMed  CAS  Google Scholar 

  2. Semenza GL (2000). Chairman’s summary: mechanisms of oxygen homeostasis, circa 1999. Adv Exp Med Biol 475:303–310

    Article  PubMed  CAS  Google Scholar 

  3. Peng YJ, Rennison J, Prabhakar NR (2004). Intermittent hypoxia augments carotid body and ventilatory response to hypoxia in neonatal rat pups. J Appl Physiol 97:2020–2025

    Article  PubMed  Google Scholar 

  4. Semenza GL (2004). Hydroxylation of HIF-1: oxygen sensing at the molecular level. Physiology (Bethesda) 19:176–182

    CAS  Google Scholar 

  5. Wang GL, Semenza GL (1993). Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J Biol Chem 268:21513–21518

    PubMed  CAS  Google Scholar 

  6. Josko J, Mazurek M (2004). Transcription factors having impact on vascular endothelial growth factor (VEGF) gene expression in angiogenesis. Med Sci Monit 10:RA89–98

    PubMed  CAS  Google Scholar 

  7. Marti HH (2004). Erythropoietin and the hypoxic brain. J Exp Biol 207:3233–3242

    Article  PubMed  CAS  Google Scholar 

  8. Nagy Z, Simon L, Bori Z (2002). Regulatory mechanisms in focal cerebral ischemia. New possibilities in neuroprotective therapy. Ideggyogy Sz 55:73–85

    PubMed  Google Scholar 

  9. Scortegagna M, Ding K, Zhang Q, Oktay Y, Bennett MJ, Bennett M, Shelton JM, Richardson JA, Moe O, Garcia JA (2005). HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner. Blood 105:3133–3140

    Article  PubMed  CAS  Google Scholar 

  10. Zelko IN, Folz RJ (2005). Extracellular superoxide dismutase functions as a major repressor of hypoxia-induced erythropoietin gene expression. Endocrinology 146:332–340

    Article  PubMed  CAS  Google Scholar 

  11. Kim JW, Tchernyshyov I, Semenza GL, Dang CV (2006). HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 3:177–185

    Article  PubMed  CAS  Google Scholar 

  12. Li F, Wang Y, Zeller KI, Potter JJ, Wonsey DR, O’Donnell KA, Kim JW, Yustein JT, Lee LA, Dang CV (2005). Myc stimulates nuclearly encoded mitochondrial genes and mitochondrial biogenesis. Mol Cell Biol 25:6225–6234

    Article  PubMed  CAS  Google Scholar 

  13. Sies H, Cadenas E (1985) Oxidative stress: damage to intact cells and organs. Philos Trans R Soc Lond B Biol Sci 311(1152):617–631

    PubMed  CAS  Google Scholar 

  14. Dawson SL, Blake MJ, Panerai RB, Potter JF (2000). Dynamic but not static cerebral autoregulation is impaired in acute ischaemic stroke. Cerebrovasc Dis 10:126–132

    Article  PubMed  CAS  Google Scholar 

  15. Liu J, Narasimhan P, Yu F, Chan PH (2005). Neuroprotection by hypoxic preconditioning involves oxidative stress-mediated expression of hypoxia-inducible factor and erythropoietin. Stroke 36:1264–1269

    Article  PubMed  CAS  Google Scholar 

  16. Mackensen GB, Patel M, Sheng H, Calvi CL, Batinic-Haberle I, Day BJ, Liang LP, Fridovich I, Crapo JD, Pearlstein RD, Warner DS (2001). Neuroprotection from delayed postischemic administration of a metalloporphyrin catalytic antioxidant. J Neurosci 21:4582–4592

    PubMed  CAS  Google Scholar 

  17. Alexandrova ML, Bochev PG (2005). Oxidative stress during the chronic phase after stroke. Free Radic Biol Med 39:297–316

    Article  PubMed  CAS  Google Scholar 

  18. Koziol JA, Feng AC (2006). On the analysis and interpretation of outcome measures in stroke clinical trials: lessons from the SAINT I study of NXY-059 for acute ischemic stroke. Stroke 37:2644–2647

    Article  PubMed  Google Scholar 

  19. Ralph GS, Parham S, Lee SR, Beard GL, Craigon MH, Ward N, White JR, Barber RD, Rayner W, Kingsman SM, et al (2004). Identification of potential stroke targets by lentiviral vector mediated overexpression of HIF-1 alpha and HIF-2 alpha in a primary neuronal model of hypoxia. J Cereb Blood Flow Metab 24:245–258

    Article  PubMed  CAS  Google Scholar 

  20. Ratan RR, Siddiq A, Aminova L, Lange PS, Langley B, Ayoub I, Gensert J, Chavez J (2004). Translation of ischemic preconditioning to the patient: prolyl hydroxylase inhibition and hypoxia inducible factor-1 as novel targets for stroke therapy. Stroke 35:2687–2689

    Article  PubMed  Google Scholar 

  21. Bergeron M, Gidday JM, Yu AY, Semenza GL, Ferriero DM, Sharp FR (2000). Role of hypoxia-inducible factor-1 in hypoxia-induced ischemic tolerance in neonatal rat brain. Ann Neurol 48:285–296

    Article  PubMed  CAS  Google Scholar 

  22. Sharp FR, Bergeron M, Bernaudin M (2001). Hypoxia-inducible factor in brain. Adv Exp Med Biol 502:273–291

    PubMed  CAS  Google Scholar 

  23. Mu D, Jiang X, Sheldon RA, Fox CK, Hamrick SE, Vexler ZS, Ferriero DM (2003). Regulation of hypoxia-inducible factor 1alpha and induction of vascular endothelial growth factor in a rat neonatal stroke model. Neurobiol Dis 14:524–534

    Article  PubMed  CAS  Google Scholar 

  24. Tupitsyna TV, Slominskii PA et al (2006) Association of the IVS9-675C > A polymorphism of the HIF-1alpha gene with acute ischemic stroke in the Moscow population. Genetika 42(6):858–861

    PubMed  CAS  Google Scholar 

  25. Zaman K, Ryu H, Hall D, O’Donovan K, Lin KI, Miller MP, Marquis JC, Baraban JM, Semenza GL, Ratan RR (1999). Protection from oxidative stress-induced apoptosis in cortical neuronal cultures by iron chelators is associated with enhanced DNA binding of hypoxia-inducible factor-1 and ATF-1/CREB and increased expression of glycolytic enzymes, p21(waf1/cip1), and erythropoietin. J Neurosci 19:9821–9830

    PubMed  CAS  Google Scholar 

  26. Bruick RK, McKnight SL (2001). A conserved family of prolyl-4-hydroxylases that modify HIF Science 294:1337–1340

    CAS  Google Scholar 

  27. Epstein AC, Gleadle JM, McNeill LA, Hewitson KS, O’Rourke J, Mole DR, Mukherji M, Metzen E, Wilson MI, Dhanda A, et al (2001). C. elegans EGL-9 and mammalian homologs define a family of dioxygenases that regulate HIF by prolyl hydroxylation. Cell 107:43–54

    Article  PubMed  CAS  Google Scholar 

  28. Hirsila M, Koivunen P, Gunzler V, Kivirikko KI, Myllyharju J (2003). Characterization of the human prolyl 4-hydroxylases that modify the hypoxia-inducible factor. J Biol Chem 278:30772–30780

    Article  PubMed  CAS  Google Scholar 

  29. Ivan M, Haberberger T, Gervasi DC, Michelson KS, Gunzler V, Kondo K, Yang H, Sorokina I, Conaway RC, Conaway JW, Kaelin Jr WG (2002). Biochemical purification and pharmacological inhibition of a mammalian prolyl hydroxylase acting on hypoxia-inducible factor. Proc Natl Acad Sci USA 99:13459–13464

    Article  PubMed  CAS  Google Scholar 

  30. Jaakkola P, Mole DR, Tian YM, Wilson MI, Gielbert J, Gaskell SJ, Kriegsheim A, Hebestreit HF, Mukherji M, Schofield CJ, et al (2001). Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292:468–472

    PubMed  CAS  Google Scholar 

  31. Siddiq A, Ayoub IA, Chavez JC, Aminova L, Shah S, LaManna JC, Patton SM, Connor JR, Cherny RA, Volitakis I, et al (2005). Hypoxia-inducible factor prolyl 4-hydroxylase inhibition. A target for neuroprotection in the central nervous system. J Biol Chem 280:41732–41743

    Article  PubMed  CAS  Google Scholar 

  32. Elkins JM, Hewitson KS et al (2003) Structure of factor-inhibiting hypoxiainducible factor (HIF) reveals mechanism of oxidative modification of HIF-1 alpha. J Biol Chem 278(3):1802–1806

    Article  PubMed  CAS  Google Scholar 

  33. Koivunen P, Hirsila M et al (2004) Catalytic properties of the asparaginyl hydroxylase (FIH) in the oxygen sensing pathway are distinct from those of its prolyl 4-hydroxylases. J Biol Chem 279(11):9899–9904

    Article  PubMed  CAS  Google Scholar 

  34. Schlemminger I, Mole DR et al (2003) Analogues of dealanylalahopcin are inhibitors of human HIF prolyl hydroxylases. Bioorg Med Chem Lett 13(8):1451–1454

    Article  PubMed  CAS  Google Scholar 

  35. Willam C, Masson N, Tian YM, Mahmood SA, Wilson MI, Bicknell R, Eckardt KU, Maxwell PH, Ratcliffe PJ, Pugh CW (2002). Peptide blockade of HIFalpha degradation modulates cellular metabolism and angiogenesis. Proc Natl Acad Sci USA 99:10423–10428

    Article  PubMed  CAS  Google Scholar 

  36. Yu F, White SB, Zhao Q, Lee FS (2001b). HIF-1alpha binding to VHL is regulated by stimulus-sensitive proline hydroxylation. Proc Natl Acad Sci USA 98:9630–9635

    Article  CAS  Google Scholar 

  37. Wenger RH, Stiehl DP, Camenisch G (2005). Integration of oxygen signaling at the consensus HRE Sci STKE 2005, re12

  38. Bruick RK (2003). Oxygen sensing in the hypoxic response pathway: regulation of the hypoxia-inducible transcription factor. Genes Dev 17:2614–2623

    Article  PubMed  CAS  Google Scholar 

  39. Hewitson KS, McNeill LA, Riordan MV, Tian YM, Bullock AN, Welford RW, Elkins JM, Oldham NJ, Bhattacharya S, Gleadle JM, et al (2002). Hypoxia-inducible factor (HIF) asparagine hydroxylase is identical to factor inhibiting HIF (FIH) and is related to the cupin structural family. J Biol Chem 277:26351–26355

    Article  PubMed  CAS  Google Scholar 

  40. Hanauske-Abel HM (1991). Prolyl 4-hydroxylase, a target enzyme for drug development. Design of suppressive agents and the in vitro effects of inhibitors and proinhibitors. J Hepatol 13(Suppl 3):S8–15; discussion S16

    Google Scholar 

  41. Hanauske-Abel HM, Gunzler V (1982). A stereochemical concept for the catalytic mechanism of prolylhydroxylase: applicability to classification and design of inhibitors. J Theor Biol 94:421–455

    Article  PubMed  CAS  Google Scholar 

  42. Lamberg A, Pihlajaniemi T, Kivirikko KI (1995). Site-directed mutagenesis of the alpha subunit of human prolyl 4-hydroxylase. Identification of three histidine residues critical for catalytic activity. J Biol Chem 270:9926–9931

    Article  PubMed  CAS  Google Scholar 

  43. Myllyharju J, Kivirikko KI (1997). Characterization of the iron- and 2-oxoglutarate-binding sites of human prolyl 4-hydroxylase. Embo J 16:1173–1180

    Article  PubMed  CAS  Google Scholar 

  44. Roach PL, Clifton IJ, Fulop V, Harlos K, Barton GJ, Hajdu J, Andersson I, Schofield CJ, Baldwin JE (1995). Crystal structure of isopenicillin N synthase is the first from a new structural family of enzymes. Nature 375:700–704

    Article  PubMed  CAS  Google Scholar 

  45. Roach PL, Clifton IJ, Hensgens CM, Shibata N, Schofield CJ, Hajdu J, Baldwin JE (1997). Structure of isopenicillin N synthase complexed with substrate and the mechanism of penicillin formation. Nature 387:827–830

    Article  PubMed  CAS  Google Scholar 

  46. Acker T, Acker H (2004). Cellular oxygen sensing need in CNS function: physiological and pathological implications. J Exp Biol 207:3171–3188

    Article  PubMed  CAS  Google Scholar 

  47. Chavez JC, LaManna JC (2002). Activation of hypoxia-inducible factor-1 in the rat cerebral cortex after transient global ischemia: potential role of insulin-like growth factor-1. J Neurosci 22:8922–8931

    PubMed  CAS  Google Scholar 

  48. Koivunen P, Hirsila M, Kivirikko KI, Myllyharju J (2006). The length of peptide substrates has a marked effect on hydroxylation by the hypoxia-inducible factor prolyl 4-hydroxylases. J Biol Chem 281:28712–28720

    Article  PubMed  CAS  Google Scholar 

  49. Stroka DM, Burkhardt T, Desbaillets I, Wenger RH, Neil DA, Bauer C, Gassmann M, Candinas D (2001). HIF-1 is expressed in normoxic tissue and displays an organ-specific regulation under systemic hypoxia. Faseb J 15:2445–2453

    PubMed  CAS  Google Scholar 

  50. Wiesener MS, Maxwell PH (2003). HIF and oxygen sensing; as important to life as the air we breathe? Ann Med 35:183–190

    Article  PubMed  CAS  Google Scholar 

  51. Koong AC, Chen EY, Mivechi NF, Denko NC, Stambrook P, Giaccia AJ (1994). Hypoxic activation of nuclear factor-kappa B is mediated by a Ras and Raf signaling pathway and does not involve MAP kinase (ERK1 or ERK2). Cancer Res 54:5273–5279

    PubMed  CAS  Google Scholar 

  52. Yan SF, Lu J, Zou YS, Soh-Won J, Cohen DM, Buttrick PM, Cooper DR, Steinberg SF, Mackman N, Pinsky DJ, Stern DM (1999). Hypoxia-associated induction of early growth response-1 gene expression. J Biol Chem 274:15030–15040

    Article  PubMed  CAS  Google Scholar 

  53. Knowles HJ, Raval RR et al (2003) Effect of ascorbate on the activity of hypoxiainducible factor in cancer cells. Cancer Res 63(8):1764–1768

    PubMed  CAS  Google Scholar 

  54. Yuan Y, Hilliard G, Ferguson T, Millhorn DE (2003). Cobalt inhibits the interaction between hypoxia-inducible factor-alpha and von Hippel-Lindau protein by direct binding to hypoxia-inducible factor-alpha. J Biol Chem 278:15911–15916

    Article  PubMed  CAS  Google Scholar 

  55. Hirsila M, Koivunen P, Xu L, Seeley T, Kivirikko KI, Myllyharju J (2005). Effect of desferrioxamine and metals on the hydroxylases in the oxygen sensing pathway. Faseb J 19:1308–1310

    PubMed  CAS  Google Scholar 

  56. Esposito BP, Epsztejn S, Breuer W, Cabantchik ZI (2002). A review of fluorescence methods for assessing labile iron in cells and biological fluids. Anal Biochem 304:1–18

    Article  PubMed  CAS  Google Scholar 

  57. Safran M, Kim WY, O’Connell F, Flippin L, Gunzler V, Horner JW, Depinho RA, Kaelin Jr WG (2006). Mouse model for noninvasive imaging of HIF prolyl hydroxylase activity: assessment of an oral agent that stimulates erythropoietin production. Proc Natl Acad Sci USA 103:105–110

    Article  PubMed  CAS  Google Scholar 

  58. Chandel NS, Maltepe E, Goldwasser E, Mathieu CE, Simon MC, Schumacker PT (1998). Mitochondrial reactive oxygen species trigger hypoxia-induced transcription. Proc Natl Acad Sci USA 95:11715–11720

    Article  PubMed  CAS  Google Scholar 

  59. Lu H, Dalgard CL, Mohyeldin A, McFate T, Tait AS, Verma A (2005). Reversible inactivation of HIF-1 prolyl hydroxylases allows cell metabolism to control basal HIF-1. J Biol Chem 280:41928–41939

    Article  PubMed  CAS  Google Scholar 

  60. Gerald D, Berra E, Frapart YM, Chan DA, Giaccia AJ, Mansuy D, Pouyssegur J, Yaniv M, Mechta-Grigoriou F (2004). JunD reduces tumor angiogenesis by protecting cells from oxidative stress. Cell 118:781–794

    Article  PubMed  CAS  Google Scholar 

  61. Myllyla R, Kuutti-Savolainen ER, Kivirikko KI (1978). The role of ascorbate in the prolyl hydroxylase reaction. Biochem Biophys Res Commun 83:441–448

    Article  PubMed  CAS  Google Scholar 

  62. Winkler P, Schaur RJ, Schauenstein E (1984). Selective promotion of ferrous ion-dependent lipid peroxidation in Ehrlich ascites tumor cells by histidine as compared with other amino acids. Biochim Biophys Acta 796:226–231

    PubMed  CAS  Google Scholar 

  63. Shoichet SA, Baumer AT, Stamenkovic D, Sauer H, Pfeiffer AF, Kahn CR, Muller-Wieland D, Richter C, Ristow M (2002). Frataxin promotes antioxidant defense in a thiol-dependent manner resulting in diminished malignant transformation in vitro. Hum Mol Genet 11:815–821

    Article  PubMed  CAS  Google Scholar 

  64. BelAiba RS, Djordjevic T, Bonello S, Flugel D, Hess J, Kietzmann T, Gorlach A (2004). Redox-sensitive regulation of the HIF pathway under non-hypoxic conditions in pulmonary artery smooth muscle cells. Biol Chem 385:249–257

    Article  PubMed  CAS  Google Scholar 

  65. Moeller BJ, Dewhirst MW (2004). Raising the bar: how HIF-1 helps determine tumor radiosensitivity. Cell Cycle 3:1107–1110

    PubMed  CAS  Google Scholar 

  66. Carrero P, Okamoto K, Coumailleau P, O’Brien S, Tanaka H, Poellinger L (2000). Redox-regulated recruitment of the transcriptional coactivators CREB-binding protein and SRC-1 to hypoxia-inducible factor 1alpha. Mol Cell Biol 20:402–415

    Article  PubMed  CAS  Google Scholar 

  67. Ema M, Hirota K, Mimura J, Abe H, Yodoi J, Sogawa K, Poellinger L, Fujii-Kuriyama Y (1999). Molecular mechanisms of transcription activation by HLF and HIF1alpha in response to hypoxia: their stabilization and redox signal-induced interaction with CBP/p300. Embo J 18:1905–1914

    Article  PubMed  CAS  Google Scholar 

  68. Huang LE, Arany Z, Livingston DM, Bunn HF (1996). Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its alpha subunit. J Biol Chem 271:32253–32259

    Article  PubMed  CAS  Google Scholar 

  69. Lando D, Pongratz I, Poellinger L, Whitelaw ML (2000). A redox mechanism controls differential DNA binding activities of hypoxia-inducible factor (HIF) 1alpha and the HIF-like factor. J Biol Chem 275:4618–4627

    Article  PubMed  CAS  Google Scholar 

  70. Welsh SJ, Bellamy WT, Briehl MM, Powis G (2002). The redox protein thioredoxin-1 (Trx-1) increases hypoxia-inducible factor 1alpha protein expression: Trx-1 overexpression results in increased vascular endothelial growth factor production and enhanced tumor angiogenesis. Cancer Res 62:5089–5095

    PubMed  CAS  Google Scholar 

  71. Mans AM, DeJoseph MR, Hawkins RA (1994). Metabolic abnormalities and grade of encephalopathy in acute hepatic failure. J Neurochem 63:1829–1838

    Article  PubMed  CAS  Google Scholar 

  72. Dalgard CL, Lu H, Mohyeldin A, Verma A (2004). Endogenous 2-oxoacids differentially regulate expression of oxygen sensors. Biochem J 380:419–424

    Article  PubMed  CAS  Google Scholar 

  73. Isaacs JS, Jung YJ, Mole DR, Lee S, Torres-Cabala C, Chung YL, Merino M, Trepel J, Zbar B, Toro J, et al (2005). HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell 8:143–153

    Article  PubMed  CAS  Google Scholar 

  74. Pollard PJ, Briere JJ, Alam NA, Barwell J, Barclay E, Wortham NC, Hunt T, Mitchell M, Olpin S, Moat SJ, et al (2005). Accumulation of Krebs cycle intermediates and over-expression of HIF1alpha in tumours which result from germline FH and SDH mutations. Hum Mol Genet 14:2231–2239

    Article  PubMed  CAS  Google Scholar 

  75. Selak MA, Armour SM, MacKenzie ED, Boulahbel H, Watson DG, Mansfield KD, Pan Y, Simon MC, Thompson CB, Gottlieb E (2005). Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell 7:77–85

    Article  PubMed  CAS  Google Scholar 

  76. Chan DA, Sutphin PD, Denko NC, Giaccia AJ (2002). Role of prolyl hydroxylation in oncogenically stabilized hypoxia-inducible factor-1alpha. J Biol Chem 277:40112–40117

    Article  PubMed  CAS  Google Scholar 

  77. Astuti D, Latif F, Dallol A, Dahia PL, Douglas F, George E, Skoldberg F, Husebye ES, Eng C, Maher ER (2001). Gene mutations in the succinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J Hum Genet 69:49–54

    Article  PubMed  CAS  Google Scholar 

  78. Baysal BE, Ferrell RE, Willett-Brozick JE, Lawrence EC, Myssiorek D, Bosch A, van der Mey A, Taschner PE, Rubinstein WS, Myers EN, et al (2000). Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 287:848–851

    Article  PubMed  CAS  Google Scholar 

  79. Niemann S, Muller U (2000). Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet 26:268–270

    Article  PubMed  CAS  Google Scholar 

  80. Pollard PJ, Wortham NC, Tomlinson IP (2003). The TCA cycle and tumorigenesis: the examples of fumarate hydratase and succinate dehydrogenase. Ann Med 35:632–639

    PubMed  CAS  Google Scholar 

  81. Eng C, Kiuru M, Fernandez MJ, Aaltonen LA (2003). A role for mitochondrial enzymes in inherited neoplasia and beyond. Nat Rev Cancer 3:193–202

    Article  PubMed  CAS  Google Scholar 

  82. Marti HH, Katschinski DM, Wagner KF, Schaffer L, Stier B, Wenger RH (2002). Isoform-specific expression of hypoxia-inducible factor-1alpha during the late stages of mouse spermiogenesis. Mol Endocrinol 16:234–243

    Article  PubMed  CAS  Google Scholar 

  83. Semenza GL (1999). Perspectives on oxygen sensing. Cell 98:281–284

    Article  PubMed  CAS  Google Scholar 

  84. Wenger RH (2002). Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression. Faseb J 16:1151–1162

    Article  PubMed  CAS  Google Scholar 

  85. Bardos JI, Ashcroft M (2005). Negative and positive regulation of HIF-1: a complex network. Biochim Biophys Acta 1755:107–120

    PubMed  CAS  Google Scholar 

  86. Carroll VA, Ashcroft M (2005). Targeting the molecular basis for tumour hypoxia. Expert Rev Mol Med 7:1–16

    Article  PubMed  Google Scholar 

  87. Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ (1999). The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399:271–275

    Article  PubMed  CAS  Google Scholar 

  88. Wiesener MS, Turley H, Allen WE, Willam C, Eckardt KU, Talks KL, Wood SM, Gatter KC, Harris AL, Pugh CW, et al (1998). Induction of endothelial PAS domain protein-1 by hypoxia: characterization and comparison with hypoxia-inducible factor-1alpha. Blood 92:2260–2268

    PubMed  CAS  Google Scholar 

  89. Flamme I, Frohlich T, von Reutern M, Kappel A, Damert A, Risau W (1997). HRF, a putative basic helix-loop-helix-PAS-domain transcription factor is closely related to hypoxia-inducible factor-1 alpha and developmentally expressed in blood vessels. Mech Dev 63:51–60

    Article  PubMed  CAS  Google Scholar 

  90. Tian H, McKnight SL, Russell DW (1997). Endothelial PAS domain protein 1 (EPAS1), a transcription factor selectively expressed in endothelial cells. Genes Dev 11:72–82

    PubMed  CAS  Google Scholar 

  91. Akeno N, Robins J, Zhang M, Czyzyk-Krzeska MF, Clemens TL (2002). Induction of vascular endothelial growth factor by IGF-I in osteoblast-like cells is mediated by the PI3K signaling pathway through the hypoxia-inducible factor-2alpha. Endocrinology 143:420–425

    Article  PubMed  CAS  Google Scholar 

  92. Jones A, Fujiyama C, Blanche C, Moore JW, Fuggle S, Cranston D, Bicknell R, Harris AL (2001). Relation of vascular endothelial growth factor production to expression and regulation of hypoxia-inducible factor-1 alpha and hypoxia-inducible factor-2 alpha in human bladder tumors and cell lines. Clin Cancer Res 7:1263–1272

    PubMed  CAS  Google Scholar 

  93. Rosenberger C, Mandriota S, Jurgensen JS, Wiesener MS, Horstrup JH, Frei U, Ratcliffe PJ, Maxwell PH, Bachmann S, Eckardt KU (2002). Expression of hypoxia-inducible factor-1alpha and -2alpha in hypoxic and ischemic rat kidneys. J Am Soc Nephrol 13:1721–1732

    Article  PubMed  CAS  Google Scholar 

  94. Sowter HM, Raval RR, Moore JW, Ratcliffe PJ, Harris AL (2003). Predominant role of hypoxia-inducible transcription factor (Hif)-1alpha versus Hif-2alpha in regulation of the transcriptional response to hypoxia. Cancer Res 63:6130–6134

    PubMed  CAS  Google Scholar 

  95. Brusselmans K, Bono F, Maxwell P, Dor Y, Dewerchin M, Collen D, Herbert JM, Carmeliet P (2001). Hypoxia-inducible factor-2alpha (HIF-2alpha) is involved in the apoptotic response to hypoglycemia but not to hypoxia. J Biol Chem 276:39192–39196

    Article  PubMed  CAS  Google Scholar 

  96. Compernolle V, Brusselmans K, Acker T, Hoet P, Tjwa M, Beck H, Plaisance S, Dor Y, Keshet E, Lupu F, et al (2002). Loss of HIF-2alpha and inhibition of VEGF impair fetal lung maturation, whereas treatment with VEGF prevents fatal respiratory distress in premature mice. Nat Med 8:702–710

    PubMed  CAS  Google Scholar 

  97. Kotch LE, Iyer NV, Laughner E, Semenza GL (1999). Defective vascularization of HIF-1alpha-null embryos is not associated with VEGF deficiency but with mesenchymal cell death. Dev Biol 209:254–267

    Article  PubMed  CAS  Google Scholar 

  98. Pugh CW, Ratcliffe PJ (2003). Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 9:677–684

    Article  PubMed  CAS  Google Scholar 

  99. Hara S, Hamada J, Kobayashi C, Kondo Y, Imura N (2001). Expression and characterization of hypoxia-inducible factor (HIF)-3alpha in human kidney: suppression of HIF-mediated gene expression by HIF-3alpha. Biochem Biophys Res Commun 287:808–813

    Article  PubMed  CAS  Google Scholar 

  100. Makino Y, Cao R, Svensson K, Bertilsson G, Asman M, Tanaka H, Cao Y, Berkenstam A, Poellinger L (2001). Inhibitory PAS domain protein is a negative regulator of hypoxia-inducible gene expression. Nature 414:550–554

    Article  PubMed  CAS  Google Scholar 

  101. Sipe JC, Lee P, Beutler E (2002). Brain iron metabolism and neurodegenerative disorders. Dev Neurosci 24:188–196

    Article  PubMed  CAS  Google Scholar 

  102. Berg D, Gerlach M, Youdim MB, Double KL, Zecca L, Riederer P, Becker G (2001). Brain iron pathways and their relevance to Parkinson’s disease. J Neurochem 79:225–236

    Article  PubMed  CAS  Google Scholar 

  103. Bishop GM, Robinson SR, Liu Q, Perry G, Atwood CS, Smith MA (2002). Iron: a pathological mediator of Alzheimer disease? Dev Neurosci 24:184–187

    Article  PubMed  CAS  Google Scholar 

  104. Ke Y, Ming Qian Z (2003). Iron misregulation in the brain: a primary cause of neurodegenerative disorders. Lancet Neurol 2:246–253

    Article  PubMed  CAS  Google Scholar 

  105. Rouault TA (2001). Systemic iron metabolism: a review and implications for brain iron metabolism. Pediatr Neurol 25:130–137

    Article  PubMed  CAS  Google Scholar 

  106. Henderson BR, Kuhn LC (1995). Differential modulation of the RNA-binding proteins IRP-1 and IRP-2 in response to iron. IRP-2 inactivation requires translation of another protein. J Biol Chem 270:20509–20515

    Article  PubMed  CAS  Google Scholar 

  107. Hanson ES, Foot LM, Leibold EA (1999). Hypoxia post-translationally activates iron-regulatory protein 2. J Biol Chem 274:5047–5052

    Article  PubMed  CAS  Google Scholar 

  108. Guo B, Phillips JD, Yu Y, Leibold EA (1995). Iron regulates the intracellular degradation of iron regulatory protein 2 by the proteasome. J Biol Chem 270:21645–21651

    Article  PubMed  CAS  Google Scholar 

  109. Hanson ES, Rawlins ML, Leibold EA (2003). Oxygen and iron regulation of iron regulatory protein 2. J Biol Chem 278:40337–40342

    Article  PubMed  CAS  Google Scholar 

  110. Schofield CJ, Zhang Z (1999). Structural and mechanistic studies on 2-oxoglutarate-dependent oxygenases and related enzymes. Curr Opin Struct Biol 9:722–731

    Article  PubMed  CAS  Google Scholar 

  111. Wang J, Chen G, Muckenthaler M, Galy B, Hentze MW, Pantopoulos K (2004). Iron-mediated degradation of IRP2, an unexpected pathway involving a 2-oxoglutarate-dependent oxygenase activity. Mol Cell Biol 24:954–965

    Article  PubMed  CAS  Google Scholar 

  112. Wang J, Pantopoulos K (2005). The pathway for IRP2 degradation involving 2-oxoglutarate-dependent oxygenase(s) does not require the E3 ubiquitin ligase activity of pVHL. Biochim Biophys Acta 1743:79–85

    Article  PubMed  CAS  Google Scholar 

  113. Yu F, White SB, Zhao Q, Lee FS (2001a). Dynamic, site-specific interaction of hypoxia-inducible factor-1alpha with the von Hippel-Lindau tumor suppressor protein. Cancer Res 61:4136–4142

    CAS  Google Scholar 

  114. Kuznetsova AV, Meller J, Schnell PO, Nash JA, Ignacak ML, Sanchez Y, Conaway JW, Conaway RC, Czyzyk-Krzeska MF (2003). von Hippel-Lindau protein binds hyperphosphorylated large subunit of RNA polymerase II through a proline hydroxylation motif and targets it for ubiquitination. Proc Natl Acad Sci USA 100:2706–2711

    Article  PubMed  CAS  Google Scholar 

  115. Lee KB, Wang D, Lippard SJ, Sharp PA (2002). Transcription-coupled and DNA damage-dependent ubiquitination of RNA polymerase II in vitro. Proc Natl Acad Sci USA 99:4239–4244

    Article  PubMed  CAS  Google Scholar 

  116. Mitsui A, Sharp PA (1999). Ubiquitination of RNA polymerase II large subunit signaled by phosphorylation of carboxyl-terminal domain. Proc Natl Acad Sci USA 96:6054–6059

    Article  PubMed  CAS  Google Scholar 

  117. Cramer P, Bushnell DA, Kornberg RD (2001). Structural basis of transcription: RNA polymerase II at 2.8 angstrom resolution. Science 292:1863–1876

    Article  PubMed  CAS  Google Scholar 

  118. Hopfer U, Hopfer H, Jablonski K, Stahl RA, Wolf G (2006). The novel WD-repeat protein Morg1 acts as a molecular scaffold for hypoxia-inducible factor prolyl hydroxylase 3 (PHD3). J Biol Chem 281:8645–8655

    Article  PubMed  CAS  Google Scholar 

  119. Lee S, Nakamura E et al (2005) Neuronal apoptosis linked to EglN3 prolyl hydroxylase and familial pheochromocytoma genes: developmental culling and cancer. Cancer Cell 8(2):155–167

    Article  PubMed  CAS  Google Scholar 

  120. D’Angelo G, Duplan E, Boyer N, Vigne P, Frelin C (2003). Hypoxia up-regulates prolyl hydroxylase activity: a feedback mechanism that limits HIF-1 responses during reoxygenation. J Biol Chem 278:38183–38187

    Article  PubMed  CAS  Google Scholar 

  121. del Peso L, Castellanos MC, Temes E, Martin-Puig S, Cuevas Y, Olmos G, Landazuri MO (2003). The von Hippel-Lindau/hypoxia-inducible factor (HIF) pathway regulates the transcription of the HIF-proline hydroxylase genes in response to low oxygen. J Biol Chem 278:48690–48695

    Article  PubMed  CAS  Google Scholar 

  122. Frew IJ, Hammond VE, Dickins RA, Quinn JM, Walkley CR, Sims NA, Schnall R, Della NG, Holloway AJ, Digby MR, et al (2003). Generation and analysis of Siah2 mutant mice. Mol Cell Biol 23:9150–9161

    Article  PubMed  CAS  Google Scholar 

  123. Nakayama K, Frew IJ, Hagensen M, Skals M, Habelhah H, Bhoumik A, Kadoya T, Erdjument-Bromage H, Tempst P, Frappell PB, et al (2004). Siah2 regulates stability of prolyl-hydroxylases, controls HIF1alpha abundance, and modulates physiological responses to hypoxia. Cell 117:941–952

    Article  PubMed  CAS  Google Scholar 

  124. Hu G, Zhang S, Vidal M, Baer JL, Xu T, Fearon ER (1997). Mammalian homologs of seven in absentia regulate DCC via the ubiquitin-proteasome pathway. Genes Dev 11:2701–2714

    PubMed  CAS  Google Scholar 

  125. Matsuzawa S, Takayama S, Froesch BA, Zapata JM, Reed JC (1998). p53-inducible human homologue of Drosophila seven in absentia (Siah) inhibits cell growth: suppression by BAG-1. Embo J 17:2736–2747

    Article  PubMed  CAS  Google Scholar 

  126. Matsuzawa SI, Reed JC (2001). Siah-1, SIP, and Ebi collaborate in a novel pathway for beta-catenin degradation linked to p53 responses. Mol Cell 7:915–926

    Article  PubMed  CAS  Google Scholar 

  127. Susini L, Passer BJ, Amzallag-Elbaz N, Juven-Gershon T, Prieur S, Privat N, Tuynder M, Gendron MC, Israel A, Amson R, et al (2001). Siah-1 binds and regulates the function of Numb. Proc Natl Acad Sci USA 98:15067–15072

    Article  PubMed  CAS  Google Scholar 

  128. Tiedt R, Bartholdy BA, Matthias G, Newell JW, Matthias P (2001). The RING finger protein Siah-1 regulates the level of the transcriptional coactivator OBF-1. Embo J 20:4143–4152

    Article  PubMed  CAS  Google Scholar 

  129. Zhang J, Guenther MG, Carthew RW, Lazar MA (1998). Proteasomal regulation of nuclear receptor corepressor-mediated repression. Genes Dev 12:1775–1780

    PubMed  CAS  Google Scholar 

  130. Nakayama K, Gazdoiu S et al (2007). Hypoxia-induced assembly of prolyl-hydroxylase, PHD3 into complexes: implications for its activity and susceptibility for degradation by the E3 ligase Siah2. Biochem J 401(1):217–226

    Article  PubMed  CAS  Google Scholar 

  131. Taylor MS (2001). Characterization and comparative analysis of the EGLN gene family. Gene 275:125–132

    Article  PubMed  CAS  Google Scholar 

  132. Lavista-Llanos S, Centanin L, Irisarri M, Russo DM, Gleadle JM, Bocca SN, Muzzopappa M, Ratcliffe PJ, Wappner P (2002). Control of the hypoxic response in Drosophila melanogaster by the basic helix-loop-helix PAS protein similar. Mol Cell Biol 22:6842–6853

    Article  PubMed  CAS  Google Scholar 

  133. Huang J, Zhao Q, Mooney SM, Lee FS (2002). Sequence determinants in hypoxia-inducible factor-1alpha for hydroxylation by the prolyl hydroxylases PHD1, PHD2, and PHD3. J Biol Chem 277:39792–39800

    Article  PubMed  CAS  Google Scholar 

  134. Metzen E, Berchner-Pfannschmidt U, Stengel P, Marxsen JH, Stolze I, Klinger M, Huang WQ, Wotzlaw C, Hellwig-Burgel T, Jelkmann W, et al (2003). Intracellular localisation of human HIF-1 alpha hydroxylases: implications for oxygen sensing. J Cell Sci 116:1319–1326

    Article  PubMed  CAS  Google Scholar 

  135. Ozer A, Wu LC, Bruick RK (2005). The candidate tumor suppressor ING4 represses activation of the hypoxia inducible factor (HIF). Proc Natl Acad Sci USA 102:7481–7486

    Article  PubMed  CAS  Google Scholar 

  136. Cioffi CL, Liu XQ, Kosinski PA, Garay M, Bowen BR (2003). Differential regulation of HIF-1 alpha prolyl-4-hydroxylase genes by hypoxia in human cardiovascular cells. Biochem Biophys Res Commun 303:947–953

    Article  PubMed  CAS  Google Scholar 

  137. Seth P, Krop I, Porter D, Polyak K (2002). Novel estrogen and tamoxifen induced genes identified by SAGE (Serial Analysis of Gene Expression). Oncogene 21:836–843

    Article  PubMed  CAS  Google Scholar 

  138. Lieb ME, Menzies K, Moschella MC, Ni R, Taubman MB (2002). Mammalian EGLN genes have distinct patterns of mRNA expression and regulation. Biochem Cell Biol 80:421–426

    Article  PubMed  CAS  Google Scholar 

  139. Oehme F, Ellinghaus P, Kolkhof P, Smith TJ, Ramakrishnan S, Hutter J, Schramm M, Flamme I (2002). Overexpression of PH-4, a novel putative proline 4-hydroxylase, modulates activity of hypoxia-inducible transcription factors. Biochem Biophys Res Commun 296:343–349

    Article  PubMed  CAS  Google Scholar 

  140. Willam C, Maxwell PH, Nichols L, Lygate C, Tian YM, Bernhardt W, Wiesener M, Ratcliffe PJ, Eckardt KU, Pugh CW (2006). HIF prolyl hydroxylases in the rat; organ distribution and changes in expression following hypoxia and coronary artery ligation. J Mol Cell Cardiol 41(1):68–77

    Article  PubMed  CAS  Google Scholar 

  141. Tian YM, Mole DR, Ratcliffe PJ, Gleadle JM (2006). Characterization of different isoforms of the HIF prolyl hydroxylase PHD1 generated by alternative initiation. Biochem J 397:179–186

    Article  PubMed  CAS  Google Scholar 

  142. Khanna S, Roy S, Maurer M, Ratan RR, Sen CK (2006) Oxygen-sensitive reset of hypoxia-inducible factor transactivation response: prolyl hydroxylases tune the biological normoxic set point. Free Radic Biol Med. 40(12):2147–2154

    Article  PubMed  CAS  Google Scholar 

  143. Aravind L, Koonin EV (2001). The DNA-repair protein AlkB, EGL-9, and leprecan define new families of 2-oxoglutarate- and iron-dependent dioxygenases. Genome Biol 2, RESEARCH0007

  144. Groulx I, Lee S (2002). Oxygen-dependent ubiquitination and degradation of hypoxia-inducible factor requires nuclear-cytoplasmic trafficking of the von Hippel-Lindau tumor suppressor protein. Mol Cell Biol 22:5319–5336

    Article  PubMed  CAS  Google Scholar 

  145. Appelhoff RJ, Tian YM, Raval RR, Turley H, Harris AL, Pugh CW, Ratcliffe PJ, Gleadle JM (2004). Differential function of the prolyl hydroxylases PHD1, PHD2, and PHD3 in the regulation of hypoxia-inducible factor. J Biol Chem 279:38458–38465

    Article  PubMed  CAS  Google Scholar 

  146. Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J (2003). HIF prolyl-hydroxylase 2 is the key oxygen sensor setting low steady-state levels of HIF-1alpha in normoxia. Embo J 22:4082–4090

    Article  PubMed  CAS  Google Scholar 

  147. Marxsen JH, Stengel P, Doege K, Heikkinen P, Jokilehto T, Wagner T, Jelkmann W, Jaakkola P, Metzen E (2004). Hypoxia-inducible factor-1 (HIF-1) promotes its degradation by induction of HIF-alpha-prolyl-4-hydroxylases. Biochem J 381:761–767

    Article  PubMed  CAS  Google Scholar 

  148. Metzen E, Stiehl DP, Doege K, Marxsen JH, Hellwig-Burgel T, Jelkmann W (2005). Regulation of the prolyl hydroxylase domain protein 2 (phd2/egln-1) gene: identification of a functional hypoxia-responsive element. Biochem J 387:711–717

    Article  PubMed  CAS  Google Scholar 

  149. Pescador N, Cuevas Y, Naranjo S, Alcaide M, Villar D, Landazuri MO, Del Peso L (2005). Identification of a functional hypoxia-responsive element that regulates the expression of the egl nine homologue 3 (egln3/phd3) gene. Biochem J 390:189–197

    Article  PubMed  CAS  Google Scholar 

  150. Chan DA, Sutphin PD, Yen SE, Giaccia AJ (2005). Coordinate regulation of the oxygen-dependent degradation domains of hypoxia-inducible factor 1 alpha. Mol Cell Biol 25:6415–6426

    Article  PubMed  CAS  Google Scholar 

  151. Cervera AM, Apostolova N, Luna-Crespo F, Sanjuan-Pla A, Garcia-Bou R, McCreath KJ (2006). An alternatively spliced transcript of the PHD3 gene retains prolyl hydroxylase activity. Cancer Lett 233:131–138

    Article  PubMed  CAS  Google Scholar 

  152. Freeman RS, Hasbani DM, Lipscomb EA, Straub JA, Xie L (2003). SM-20, EGL-9, and the EGLN family of hypoxia-inducible factor prolyl hydroxylases. Mol Cells 16:1–12

    PubMed  CAS  Google Scholar 

  153. Wax SD, Rosenfield CL, Taubman MB (1994). Identification of a novel growth factor-responsive gene in vascular smooth muscle cells. J Biol Chem 269:13041–13047

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ambreena Siddiq.

Additional information

Special issue dedicated to John P. Blass.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Siddiq, A., Aminova, L.R. & Ratan, R.R. Hypoxia Inducible Factor Prolyl 4-Hydroxylase Enzymes: Center Stage in the Battle Against Hypoxia, Metabolic Compromise and Oxidative Stress. Neurochem Res 32, 931–946 (2007). https://doi.org/10.1007/s11064-006-9268-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-006-9268-7

Keywords

Navigation