TP53 Regulates Metabolic Genes (Homo sapiens)

From WikiPathways

Jump to: navigation, search
6, 8, 11, 15, 17...29, 39, 69, 9323, 46, 5552, 7336987, 8859, 9754, 57, 916, 118, 58, 74, 8055, 76454, 8, 58, 8037, 8235, 79, 8931, 33, 38, 83, 849437, 827, 532, 10, 19, 20, 24...9, 38, 8635, 426025, 4547, 56, 77721, 48, 8511, 63, 7859, 715, 26, 30, 36, 50...7249, 65mitochondrial matrixcytosolnucleoplasmmitochondrial intermembrane spacelysosomal lumenAGO2 SESN3 SESN3 YWHAG PimiR-26A RISCp-S939,T1462-TSC2GLS2 dimerTSC1:TSC2EIF2C3 p-S939,T1462-TSC2 p-T,p-S-AKTSESN2 GPI TP53 Tetramer:PTENGeneCOX14 PRKAG1 PRKAG3 YWHAE DDIT4 YWHAQ TSC1:p-S1387-TSC2PRKAB2 COX11,14,16,18,20COX8A p-T308,S473-AKT1 GSHferroheme LAMTOR3 SCO2H+RPTOR RRAGC COX ancilliaryproteinsLAMTOR4 NADP+EIF2C3 TACO1 TP53 Tetramer:SCO2GeneCOX6A1 p-S939,T1462-TSC2:14-3-3 dimerYWHAQ YWHAH EIF2C4 O2MOV10 GPI dimerH+Respiratory electrontransport, ATPsynthesis bychemiosmoticcoupling, and heatproduction byuncouplingproteins.GLS2 GenePRKAG2 D-Glucono-1,5-lactone 6-phosphateG6PMLST8 SESN2 Gene p-T305,S472-AKT3 H+COX11 SCO2 Gene TP53 TSC2TSC2 2xHC-TXNmiR-26A1 DDIT4 GeneL-GlnH2O2COX4I1 YWHAB PRDX1,2,5COX18 LAMTOR1 p-T172-PRKAA2 H2OTNRC6B Energy dependentregulation of mTORby LKB1-AMPKSFN YWHAH H2ONADP+ COX19AGO2 TP53 Tetramer:RRM2BGeneYWHAB SESN1,2,3:p-AMPKheterotrimer:AMPGLS dimersSESN2 Gene ATPPTEN mRNA:miR-26ARISCH+ADPTP53 YWHAZ GTP SESN1-1,SESN1-3 TP63 TNRC6C PRDX1 miR-26A2 TP63 MTOR HOOS-C52-PRDX1 dimerYWHAZ DDIT4SESN3 Gene p-S1387-TSC2 SESN1 Gene YWHAE YWHAQ TNRC6B NH4+PRDX1 TSC1 COX6C(3-75) COX6B1 Cytochrome c oxidaseTIGARLAMTOR5 PRKAB1 ferriheme MT-CO1 GSSGTIGAR Gene G6PD LAMTOR2 GTP PRDX1 EIF2C1 SESN1 Gene TSC1TP53 GLS2 L-GluGSR-2 LAMTOR5 L-selenocysteine-residue-GPX2 COX20 SLC38A9 TP63 Tetramer/ TP53TetramerRPTOR SESN1,2,3 GenesLAMTOR4 COX5A SURF1 SESN1-1,SESN1-3 H2ORRAGC PRDX2 TP53 mTORC1:Ragulator:Rag:GNP:RHEB:GDPCOX16 MLST8 Active mTORC1complexGLS2 MT-CO2 PIP3 activates AKTsignalingMT-CO3 Metabolism ofcarbohydratesSLC38A9 HOOS-C52-PRDX1 p-AMPKheterotrimer:AMPTP53 Tetramer:TIGARGeneSFN Metabolism ofnucleotidesp-T172-PRKAA2 PRKAG2 YWHAB LRPPRC YWHAG Detoxification ofReactive OxygenSpeciesSFN TP63/T53:DDIT4 GeneTP53 TetramerNDUFA4 p-T309,S474-AKT2 RRM2BTP53Tetramer:SESN1,2,3GenesTP53 SESN1,2,3:HOOS-C52-PRDX1 dimerSCO2 GeneTP53 Fru(6)PGDP EIF2C1 DDIT4 Gene FAD GPX2 tetramerPRDX5 LAMTOR3 SESN2 NADP+SESN3 Gene TNRC6A TP53 p-T183-PRKAA1 SESN1,2,3PTEN geneGLS2 Gene SESN1-1,SESN1-3 RHEB TP53 G6PD dimer andtetramerHOOS-C52-PRDX1 Cytochrome c(oxidised)CYCS ADPGSR-2:FAD dimerMetabolism of aminoacids andderivativesTP53 PRKAB1 PRKAG1 AMP ATPMTOR miR-26A2 TP53 Tetramer:GLS2GeneSCO2 RRAGA GLS RHEB COX7B Cytochrome c(reduced)TIGAR GeneNADPHPTENYWHAZ TNRC6A RRAGD LAMTOR1 MOV10 YWHAH YWHAE AMP RRM2B GeneRRAGB PRKAB2 GDP COX7C EIF2C4 14-3-3 dimerSESN2 miR-26A1 SCO1 RRAGA RRAGB TXNRD1 PTEN mRNA TXNCOX5B RRM2B Gene D-Fructose2,6-bisphosphateCYCS SESN3 PiPRKAG3 NADPHDDIT4:14-3-3 dimerTNRC6C PRDX1 H2OCOX7A2L TSC1 YWHAG CuA PTEN mRNAH2Op-T183-PRKAA1 PTEN gene PRDX1 dimerTNXRD1:FAD dimerH2OFAD RRAGD LAMTOR2 389013, 16, 22, 34, 41...4512, 28, 991435, 727918, 40, 4451, 64, 671135, 42698, 58, 805559, 9737


Description

While the p53 tumor suppressor protein (TP53) is known to inhibit cell growth by inducing apoptosis, senescence and cell cycle arrest, recent studies have found that p53 is also able to influence cell metabolism to prevent tumor development. TP53 regulates transcription of many genes involved in the metabolism of carbohydrates, nucleotides and amino acids, protein synthesis and aerobic respiration.

TP53 stimulates transcription of TIGAR, a D-fructose 2,6-bisphosphatase. TIGAR activity decreases glycolytic rate and lowers ROS (reactive oxygen species) levels in cells (Bensaad et al. 2006). TP53 may also negatively regulate the rate of glycolysis by inhibiting the expression of glucose transporters GLUT1, GLUT3 and GLUT4 (Kondoh et al. 2005, Schwartzenberg-Bar-Yoseph et al. 2004, Kawauchi et al. 2008).<p>TP53 negatively regulates several key points in PI3K/AKT signaling and downstream mTOR signaling, decreasing the rate of protein synthesis and, hence, cellular growth. TP53 directly stimulates transcription of the tumor suppressor PTEN, which acts to inhibit PI3K-mediated activation of AKT (Stambolic et al. 2001). TP53 stimulates transcription of sestrin genes, SESN1, SESN2, and SESN3 (Velasco-Miguel et al. 1999, Budanov et al. 2002, Brynczka et al. 2007). One of sestrin functions may be to reduce and reactivate overoxidized peroxiredoxin PRDX1, thereby reducing ROS levels (Budanov et al. 2004, Papadia et al. 2008, Essler et al. 2009). Another function of sestrins is to bind the activated AMPK complex and protect it from AKT-mediated inactivation. By enhancing AMPK activity, sestrins negatively regulate mTOR signaling (Budanov and Karin 2008, Cam et al. 2014). The expression of DDIT4 (REDD1), another negative regulator of mTOR signaling, is directly stimulated by TP63 and TP53. DDIT4 prevents AKT-mediated inactivation of TSC1:TSC2 complex, thus inhibiting mTOR cascade (Cam et al. 2014, Ellisen et al. 2002, DeYoung et al. 2008). TP53 may also be involved, directly or indirectly, in regulation of expression of other participants of PI3K/AKT/mTOR signaling, such as PIK3CA (Singh et al. 2002), TSC2 and AMPKB (Feng et al. 2007). <p>TP53 regulates mitochondrial metabolism through several routes. TP53 stimulates transcription of SCO2 gene, which encodes a mitochondrial cytochrome c oxidase assembly protein (Matoba et al. 2006). TP53 stimulates transcription of RRM2B gene, which encodes a subunit of the ribonucleotide reductase complex, responsible for the conversion of ribonucleotides to deoxyribonucleotides and essential for the maintenance of mitochondrial DNA content in the cell (Tanaka et al. 2000, Bourdon et al. 2007, Kulawiec et al. 2009). TP53 also transactivates mitochondrial transcription factor A (TFAM), a nuclear-encoded gene important for mitochondrial DNA (mtDNA) transcription and maintenance (Park et al. 2009). Finally, TP53 stimulates transcription of the mitochondrial glutaminase GLS2, leading to increased mitochondrial respiration rate and reduced ROS levels (Hu et al. 2010). <p>The great majority of tumor cells generate energy through aerobic glycolysis, rather than the much more efficient aerobic mitochondrial respiration, and this metabolic change is known as the Warburg effect (Warburg 1956). Since the majority of tumor cells have impaired TP53 function, and TP53 regulates a number of genes involved in glycolysis and mitochondrial respiration, it is likely that TP53 inactivation plays an important role in the metabolic derangement of cancer cells such as the Warburg effect and the concomitant increased tumorigenicity (reviewed by Feng and Levine 2010). On the other hand, some mutations of TP53 in Li-Fraumeni syndrome may result in the retention of its wild-type metabolic activities while losing cell cycle and apoptosis functions (Wang et al. 2013). Consistent with such human data, some mutations of p53, unlike p53 null state, retain the ability to regulate energy metabolism while being inactive in regulating its classic gene targets involved in cell cycle, apoptosis and senescence. Retention of metabolic and antioxidant functions of p53 protects p53 mutant mice from early onset tumorigenesis (Li et al. 2012). View original pathway at Reactome.</div>

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 5628897
Reactome-version 
Reactome version: 75
Reactome Author 
Reactome Author: Orlic-Milacic, Marija

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Elgadi KM, Meguid RA, Qian M, Souba WW, Abcouwer SF.; ''Cloning and analysis of unique human glutaminase isoforms generated by tissue-specific alternative splicing.''; PubMed Europe PMC Scholia
  2. Tian WN, Braunstein LD, Apse K, Pang J, Rose M, Tian X, Stanton RC.; ''Importance of glucose-6-phosphate dehydrogenase activity in cell death.''; PubMed Europe PMC Scholia
  3. Chu FF, Doroshow JH, Esworthy RS.; ''Expression, characterization, and tissue distribution of a new cellular selenium-dependent glutathione peroxidase, GSHPx-GI.''; PubMed Europe PMC Scholia
  4. Feng Z.; ''p53 regulation of the IGF-1/AKT/mTOR pathways and the endosomal compartment.''; PubMed Europe PMC Scholia
  5. Fontanesi F, Soto IC, Horn D, Barrientos A.; ''Assembly of mitochondrial cytochrome c-oxidase, a complicated and highly regulated cellular process.''; PubMed Europe PMC Scholia
  6. WARBURG O.; ''On the origin of cancer cells.''; PubMed Europe PMC Scholia
  7. Loos H, Roos D, Weening R, Houwerzijl J.; ''Familial deficiency of glutathione reductase in human blood cells.''; PubMed Europe PMC Scholia
  8. Velasco-Miguel S, Buckbinder L, Jean P, Gelbert L, Talbott R, Laidlaw J, Seizinger B, Kley N.; ''PA26, a novel target of the p53 tumor suppressor and member of the GADD family of DNA damage and growth arrest inducible genes.''; PubMed Europe PMC Scholia
  9. Yang KS, Kang SW, Woo HA, Hwang SC, Chae HZ, Kim K, Rhee SG.; ''Inactivation of human peroxiredoxin I during catalysis as the result of the oxidation of the catalytic site cysteine to cysteine-sulfinic acid.''; PubMed Europe PMC Scholia
  10. Boada J, Roig T, Perez X, Gamez A, Bartrons R, Cascante M, Bermúdez J.; ''Cells overexpressing fructose-2,6-bisphosphatase showed enhanced pentose phosphate pathway flux and resistance to oxidative stress.''; PubMed Europe PMC Scholia
  11. Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova O, Hurley PJ, Bunz F, Hwang PM.; ''p53 regulates mitochondrial respiration.''; PubMed Europe PMC Scholia
  12. Felig P.; ''Amino acid metabolism in man.''; PubMed Europe PMC Scholia
  13. Fukai T, Ushio-Fukai M.; ''Superoxide dismutases: role in redox signaling, vascular function, and diseases.''; PubMed Europe PMC Scholia
  14. Dashty M.; ''A quick look at biochemistry: carbohydrate metabolism.''; PubMed Europe PMC Scholia
  15. Feng Z, Hu W, de Stanchina E, Teresky AK, Jin S, Lowe S, Levine AJ.; ''The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways.''; PubMed Europe PMC Scholia
  16. Veal E, Day A.; ''Hydrogen peroxide as a signaling molecule.''; PubMed Europe PMC Scholia
  17. Park JY, Wang PY, Matsumoto T, Sung HJ, Ma W, Choi JW, Anderson SA, Leary SC, Balaban RS, Kang JG, Hwang PM.; ''p53 improves aerobic exercise capacity and augments skeletal muscle mitochondrial DNA content.''; PubMed Europe PMC Scholia
  18. Hardie DG.; ''AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy.''; PubMed Europe PMC Scholia
  19. Kletzien RF, Harris PK, Foellmi LA.; ''Glucose-6-phosphate dehydrogenase: a "housekeeping" enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress.''; PubMed Europe PMC Scholia
  20. Fico A, Paglialunga F, Cigliano L, Abrescia P, Verde P, Martini G, Iaccarino I, Filosa S.; ''Glucose-6-phosphate dehydrogenase plays a crucial role in protection from redox-stress-induced apoptosis.''; PubMed Europe PMC Scholia
  21. Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E.; ''The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression.''; PubMed Europe PMC Scholia
  22. Presnell CE, Bhatti G, Numan LS, Lerche M, Alkhateeb SK, Ghalib M, Shammaa M, Kavdia M.; ''Computational insights into the role of glutathione in oxidative stress.''; PubMed Europe PMC Scholia
  23. Kulawiec M, Ayyasamy V, Singh KK.; ''p53 regulates mtDNA copy number and mitocheckpoint pathway.''; PubMed Europe PMC Scholia
  24. Vander Heiden MG, Plas DR, Rathmell JC, Fox CJ, Harris MH, Thompson CB.; ''Growth factors can influence cell growth and survival through effects on glucose metabolism.''; PubMed Europe PMC Scholia
  25. Singh B, Reddy PG, Goberdhan A, Walsh C, Dao S, Ngai I, Chou TC, O-Charoenrat P, Levine AJ, Rao PH, Stoffel A.; ''p53 regulates cell survival by inhibiting PIK3CA in squamous cell carcinomas.''; PubMed Europe PMC Scholia
  26. Stiburek L, Hansikova H, Tesarova M, Cerna L, Zeman J.; ''Biogenesis of eukaryotic cytochrome c oxidase.''; PubMed Europe PMC Scholia
  27. Li T, Kon N, Jiang L, Tan M, Ludwig T, Zhao Y, Baer R, Gu W.; ''Tumor suppression in the absence of p53-mediated cell-cycle arrest, apoptosis, and senescence.''; PubMed Europe PMC Scholia
  28. Häussinger D.; ''Liver glutamine metabolism.''; PubMed Europe PMC Scholia
  29. Venkatanarayan A, Raulji P, Norton W, Flores ER.; ''Novel therapeutic interventions for p53-altered tumors through manipulation of its family members, p63 and p73.''; PubMed Europe PMC Scholia
  30. Schultz BE, Chan SI.; ''Structures and proton-pumping strategies of mitochondrial respiratory enzymes.''; PubMed Europe PMC Scholia
  31. Essler S, Dehne N, Brüne B.; ''Role of sestrin2 in peroxide signaling in macrophages.''; PubMed Europe PMC Scholia
  32. Kawauchi K, Araki K, Tobiume K, Tanaka N.; ''p53 regulates glucose metabolism through an IKK-NF-kappaB pathway and inhibits cell transformation.''; PubMed Europe PMC Scholia
  33. Woo HA, Bae SH, Park S, Rhee SG.; ''Sestrin 2 is not a reductase for cysteine sulfinic acid of peroxiredoxins.''; PubMed Europe PMC Scholia
  34. Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O.; ''Oxidative stress and antioxidant defense.''; PubMed Europe PMC Scholia
  35. Cam M, Bid HK, Xiao L, Zambetti GP, Houghton PJ, Cam H.; ''p53/TAp63 and AKT regulate mammalian target of rapamycin complex 1 (mTORC1) signaling through two independent parallel pathways in the presence of DNA damage.''; PubMed Europe PMC Scholia
  36. Trumpower BL, Gennis RB.; ''Energy transduction by cytochrome complexes in mitochondrial and bacterial respiration: the enzymology of coupling electron transfer reactions to transmembrane proton translocation.''; PubMed Europe PMC Scholia
  37. Hu W, Zhang C, Wu R, Sun Y, Levine A, Feng Z.; ''Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function.''; PubMed Europe PMC Scholia
  38. Budanov AV, Sablina AA, Feinstein E, Koonin EV, Chumakov PM.; ''Regeneration of peroxiredoxins by p53-regulated sestrins, homologs of bacterial AhpD.''; PubMed Europe PMC Scholia
  39. Jen KY, Cheung VG.; ''Identification of novel p53 target genes in ionizing radiation response.''; PubMed Europe PMC Scholia
  40. Katajisto P, Vallenius T, Vaahtomeri K, Ekman N, Udd L, Tiainen M, Mäkelä TP.; ''The LKB1 tumor suppressor kinase in human disease.''; PubMed Europe PMC Scholia
  41. Kavdia M.; ''Mathematical and computational models of oxidative and nitrosative stress.''; PubMed Europe PMC Scholia
  42. DeYoung MP, Horak P, Sofer A, Sgroi D, Ellisen LW.; ''Hypoxia regulates TSC1/2-mTOR signaling and tumor suppression through REDD1-mediated 14-3-3 shuttling.''; PubMed Europe PMC Scholia
  43. Brand MD.; ''The sites and topology of mitochondrial superoxide production.''; PubMed Europe PMC Scholia
  44. Guertin DA, Sabatini DM.; ''Defining the role of mTOR in cancer.''; PubMed Europe PMC Scholia
  45. Stambolic V, MacPherson D, Sas D, Lin Y, Snow B, Jang Y, Benchimol S, Mak TW.; ''Regulation of PTEN transcription by p53.''; PubMed Europe PMC Scholia
  46. Bourdon A, Minai L, Serre V, Jais JP, Sarzi E, Aubert S, Chrétien D, de Lonlay P, Paquis-Flucklinger V, Arakawa H, Nakamura Y, Munnich A, Rötig A.; ''Mutation of RRM2B, encoding p53-controlled ribonucleotide reductase (p53R2), causes severe mitochondrial DNA depletion.''; PubMed Europe PMC Scholia
  47. Zhang C, Zhang Z, Zhu Y, Qin S.; ''Glucose-6-phosphate dehydrogenase: a biomarker and potential therapeutic target for cancer.''; PubMed Europe PMC Scholia
  48. Quesada AR, Sanchez-Jimenez F, Perez-Rodriguez J, Marquez J, Medina MA, Nuñez de Castro I.; ''Purification of phosphate-dependent glutaminase from isolated mitochondria of Ehrlich ascites-tumour cells.''; PubMed Europe PMC Scholia
  49. Xu W, Beutler E.; ''The characterization of gene mutations for human glucose phosphate isomerase deficiency associated with chronic hemolytic anemia.''; PubMed Europe PMC Scholia
  50. Wikstrom MK.; ''Proton pump coupled to cytochrome c oxidase in mitochondria.''; PubMed Europe PMC Scholia
  51. Hyde JE.; ''Targeting purine and pyrimidine metabolism in human apicomplexan parasites.''; PubMed Europe PMC Scholia
  52. Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC.; ''Identification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway.''; PubMed Europe PMC Scholia
  53. SCOTT EM, DUNCAN IW, EKSTRAND V.; ''PURIFICATION AND PROPERTIES OF GLUTATHIONE REDUCTASE OF HUMAN ERYTHROCYTES.''; PubMed Europe PMC Scholia
  54. Nagy P, Karton A, Betz A, Peskin AV, Pace P, O'Reilly RJ, Hampton MB, Radom L, Winterbourn CC.; ''Model for the exceptional reactivity of peroxiredoxins 2 and 3 with hydrogen peroxide: a kinetic and computational study.''; PubMed Europe PMC Scholia
  55. Tanaka H, Arakawa H, Yamaguchi T, Shiraishi K, Fukuda S, Matsui K, Takei Y, Nakamura Y.; ''A ribonucleotide reductase gene involved in a p53-dependent cell-cycle checkpoint for DNA damage.''; PubMed Europe PMC Scholia
  56. Luzzatto L, Afolayan A.; ''Enzymic properties of different types of human erythrocyte glucose-6-phosphate dehydrogenase, with characterization of two new genetic variants.''; PubMed Europe PMC Scholia
  57. Yamashita H, Avraham S, Jiang S, London R, Van Veldhoven PP, Subramani S, Rogers RA, Avraham H.; ''Characterization of human and murine PMP20 peroxisomal proteins that exhibit antioxidant activity in vitro.''; PubMed Europe PMC Scholia
  58. Brynczka C, Labhart P, Merrick BA.; ''NGF-mediated transcriptional targets of p53 in PC12 neuronal differentiation.''; PubMed Europe PMC Scholia
  59. Cai SL, Tee AR, Short JD, Bergeron JM, Kim J, Shen J, Guo R, Johnson CL, Kiguchi K, Walker CL.; ''Activity of TSC2 is inhibited by AKT-mediated phosphorylation and membrane partitioning.''; PubMed Europe PMC Scholia
  60. Inoki K, Zhu T, Guan KL.; ''TSC2 mediates cellular energy response to control cell growth and survival.''; PubMed Europe PMC Scholia
  61. Murphy MP.; ''How mitochondria produce reactive oxygen species.''; PubMed Europe PMC Scholia
  62. Kondoh H, Lleonart ME, Gil J, Wang J, Degan P, Peters G, Martinez D, Carnero A, Beach D.; ''Glycolytic enzymes can modulate cellular life span.''; PubMed Europe PMC Scholia
  63. Qi Z, He J, Su Y, He Q, Liu J, Yu L, Al-Attas O, Hussain T, Ding S, Ji L, Qian M.; ''Physical exercise regulates p53 activity targeting SCO2 and increases mitochondrial COX biogenesis in cardiac muscle with age.''; PubMed Europe PMC Scholia
  64. Welin M, Nordlund P.; ''Understanding specificity in metabolic pathways--structural biology of human nucleotide metabolism.''; PubMed Europe PMC Scholia
  65. TSUBOI KK, ESTRADA J, HUDSON PB.; ''Enzymes of the human erythrocyte. IV. Phosphoglucose isomerase, purification and properties.''; PubMed Europe PMC Scholia
  66. Feng Z, Levine AJ.; ''The regulation of energy metabolism and the IGF-1/mTOR pathways by the p53 protein.''; PubMed Europe PMC Scholia
  67. Rudolph FB.; ''The biochemistry and physiology of nucleotides.''; PubMed Europe PMC Scholia
  68. Buettner GR.; ''Superoxide dismutase in redox biology: the roles of superoxide and hydrogen peroxide.''; PubMed Europe PMC Scholia
  69. Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E, Vousden KH.; ''TIGAR, a p53-inducible regulator of glycolysis and apoptosis.''; PubMed Europe PMC Scholia
  70. Cui G, Park S, Badeaux AI, Kim D, Lee J, Thompson JR, Yan F, Kaneko S, Yuan Z, Botuyan MV, Bedford MT, Cheng JQ, Mer G.; ''PHF20 is an effector protein of p53 double lysine methylation that stabilizes and activates p53.''; PubMed Europe PMC Scholia
  71. Fisher TL, White MF.; ''Signaling pathways: the benefits of good communication.''; PubMed Europe PMC Scholia
  72. Ellisen LW, Ramsayer KD, Johannessen CM, Yang A, Beppu H, Minda K, Oliner JD, McKeon F, Haber DA.; ''REDD1, a developmentally regulated transcriptional target of p63 and p53, links p63 to regulation of reactive oxygen species.''; PubMed Europe PMC Scholia
  73. Inoki K, Li Y, Zhu T, Wu J, Guan KL.; ''TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling.''; PubMed Europe PMC Scholia
  74. Menendez D, Nguyen TA, Freudenberg JM, Mathew VJ, Anderson CW, Jothi R, Resnick MA.; ''Diverse stresses dramatically alter genome-wide p53 binding and transactivation landscape in human cancer cells.''; PubMed Europe PMC Scholia
  75. Soto IC, Fontanesi F, Liu J, Barrientos A.; ''Biogenesis and assembly of eukaryotic cytochrome c oxidase catalytic core.''; PubMed Europe PMC Scholia
  76. Nakano K, Bálint E, Ashcroft M, Vousden KH.; ''A ribonucleotide reductase gene is a transcriptional target of p53 and p73.''; PubMed Europe PMC Scholia
  77. Au SW, Gover S, Lam VM, Adams MJ.; ''Human glucose-6-phosphate dehydrogenase: the crystal structure reveals a structural NADP(+) molecule and provides insights into enzyme deficiency.''; PubMed Europe PMC Scholia
  78. Wei CL, Wu Q, Vega VB, Chiu KP, Ng P, Zhang T, Shahab A, Yong HC, Fu Y, Weng Z, Liu J, Zhao XD, Chew JL, Lee YL, Kuznetsov VA, Sung WK, Miller LD, Lim B, Liu ET, Yu Q, Ng HH, Ruan Y.; ''A global map of p53 transcription-factor binding sites in the human genome.''; PubMed Europe PMC Scholia
  79. Budanov AV, Karin M.; ''p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling.''; PubMed Europe PMC Scholia
  80. Budanov AV, Shoshani T, Faerman A, Zelin E, Kamer I, Kalinski H, Gorodin S, Fishman A, Chajut A, Einat P, Skaliter R, Gudkov AV, Chumakov PM, Feinstein E.; ''Identification of a novel stress-responsive gene Hi95 involved in regulation of cell viability.''; PubMed Europe PMC Scholia
  81. Balsa E, Marco R, Perales-Clemente E, Szklarczyk R, Calvo E, Landázuri MO, Enríquez JA.; ''NDUFA4 is a subunit of complex IV of the mammalian electron transport chain.''; PubMed Europe PMC Scholia
  82. Suzuki S, Tanaka T, Poyurovsky MV, Nagano H, Mayama T, Ohkubo S, Lokshin M, Hosokawa H, Nakayama T, Suzuki Y, Sugano S, Sato E, Nagao T, Yokote K, Tatsuno I, Prives C.; ''Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species.''; PubMed Europe PMC Scholia
  83. Papadia S, Soriano FX, Léveillé F, Martel MA, Dakin KA, Hansen HH, Kaindl A, Sifringer M, Fowler J, Stefovska V, McKenzie G, Craigon M, Corriveau R, Ghazal P, Horsburgh K, Yankner BA, Wyllie DJ, Ikonomidou C, Hardingham GE.; ''Synaptic NMDA receptor activity boosts intrinsic antioxidant defenses.''; PubMed Europe PMC Scholia
  84. Gong F, Hou G, Liu H, Zhang M.; ''Peroxiredoxin 1 promotes tumorigenesis through regulating the activity of mTOR/p70S6K pathway in esophageal squamous cell carcinoma.''; PubMed Europe PMC Scholia
  85. Gómez-Fabre PM, Aledo JC, Del Castillo-Olivares A, Alonso FJ, Núñez De Castro I, Campos JA, Márquez J.; ''Molecular cloning, sequencing and expression studies of the human breast cancer cell glutaminase.''; PubMed Europe PMC Scholia
  86. Wood ZA, Poole LB, Karplus PA.; ''Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling.''; PubMed Europe PMC Scholia
  87. Inoki K, Li Y, Xu T, Guan KL.; ''Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling.''; PubMed Europe PMC Scholia
  88. Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J.; ''Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb.''; PubMed Europe PMC Scholia
  89. Sanli T, Linher-Melville K, Tsakiridis T, Singh G.; ''Sestrin2 modulates AMPK subunit expression and its response to ionizing radiation in breast cancer cells.''; PubMed Europe PMC Scholia
  90. Pontarin G, Fijolek A, Pizzo P, Ferraro P, Rampazzo C, Pozzan T, Thelander L, Reichard PA, Bianchi V.; ''Ribonucleotide reduction is a cytosolic process in mammalian cells independently of DNA damage.''; PubMed Europe PMC Scholia
  91. Lee W, Choi KS, Riddell J, Ip C, Ghosh D, Park JH, Park YM.; ''Human peroxiredoxin 1 and 2 are not duplicate proteins: the unique presence of CYS83 in Prx1 underscores the structural and functional differences between Prx1 and Prx2.''; PubMed Europe PMC Scholia
  92. Pitceathly RD, Rahman S, Wedatilake Y, Polke JM, Cirak S, Foley AR, Sailer A, Hurles ME, Stalker J, Hargreaves I, Woodward CE, Sweeney MG, Muntoni F, Houlden H, Taanman JW, Hanna MG, UK10K Consortium.; ''NDUFA4 mutations underlie dysfunction of a cytochrome c oxidase subunit linked to human neurological disease.''; PubMed Europe PMC Scholia
  93. Lee P, Vousden KH, Cheung EC.; ''TIGAR, TIGAR, burning bright.''; PubMed Europe PMC Scholia
  94. Urig S, Lieske J, Fritz-Wolf K, Irmler A, Becker K.; ''Truncated mutants of human thioredoxin reductase 1 do not exhibit glutathione reductase activity.''; PubMed Europe PMC Scholia
  95. Wang PY, Ma W, Park JY, Celi FS, Arena R, Choi JW, Ali QA, Tripodi DJ, Zhuang J, Lago CU, Strong LC, Talagala SL, Balaban RS, Kang JG, Hwang PM.; ''Increased oxidative metabolism in the Li-Fraumeni syndrome.''; PubMed Europe PMC Scholia
  96. Perez JX, Roig T, Manzano A, Dalmau M, Boada J, Ventura F, Rosa JL, Bermudez J, Bartrons R.; ''Overexpression of fructose 2,6-bisphosphatase decreases glycolysis and delays cell cycle progression.''; PubMed Europe PMC Scholia
  97. Liu MY, Cai S, Espejo A, Bedford MT, Walker CL.; ''14-3-3 interacts with the tumor suppressor tuberin at Akt phosphorylation site(s).''; PubMed Europe PMC Scholia
  98. Ray PD, Huang BW, Tsuji Y.; ''Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling.''; PubMed Europe PMC Scholia
  99. Owen OE, Reichard GA, Patel MS, Boden G.; ''Energy metabolism in feasting and fasting.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114836view16:33, 25 January 2021ReactomeTeamReactome version 75
113282view11:35, 2 November 2020ReactomeTeamReactome version 74
112493view15:45, 9 October 2020ReactomeTeamReactome version 73
102617view08:09, 13 January 2019EgonwAssuming CuA is Cu2+
101405view11:29, 1 November 2018ReactomeTeamreactome version 66
100943view21:05, 31 October 2018ReactomeTeamreactome version 65
100480view19:39, 31 October 2018ReactomeTeamreactome version 64
100025view16:22, 31 October 2018ReactomeTeamreactome version 63
99578view14:55, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99200view12:43, 31 October 2018ReactomeTeamreactome version 62
93881view13:42, 16 August 2017ReactomeTeamreactome version 61
93449view11:23, 9 August 2017ReactomeTeamreactome version 61
88395view15:18, 4 August 2016FehrhartOntology Term : 'classic metabolic pathway' added !
88394view15:16, 4 August 2016FehrhartOntology Term : 'regulatory pathway' added !
86541view09:20, 11 July 2016ReactomeTeamreactome version 56
83329view10:48, 18 November 2015ReactomeTeamVersion54
81482view13:01, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
14-3-3 dimerComplexR-HSA-1445138 (Reactome)
2xHC-TXNProteinP10599 (Uniprot-TrEMBL)
ADPMetaboliteCHEBI:456216 (ChEBI)
AGO2 ProteinQ9UKV8 (Uniprot-TrEMBL)
AMP MetaboliteCHEBI:16027 (ChEBI)
ATPMetaboliteCHEBI:30616 (ChEBI)
Active mTORC1 complexComplexR-HSA-165678 (Reactome)
COX ancilliary proteinsComplexR-HSA-5566488 (Reactome)
COX11 ProteinQ9Y6N1 (Uniprot-TrEMBL)
COX11,14,16,18,20ComplexR-HSA-5336143 (Reactome)
COX14 ProteinQ96I36 (Uniprot-TrEMBL)
COX16 ProteinQ9P0S2 (Uniprot-TrEMBL)
COX18 ProteinQ8N8Q8 (Uniprot-TrEMBL)
COX19ProteinQ49B96 (Uniprot-TrEMBL)
COX20 ProteinQ5RI15 (Uniprot-TrEMBL)
COX4I1 ProteinP13073 (Uniprot-TrEMBL)
COX5A ProteinP20674 (Uniprot-TrEMBL)
COX5B ProteinP10606 (Uniprot-TrEMBL)
COX6A1 ProteinP12074 (Uniprot-TrEMBL)
COX6B1 ProteinP14854 (Uniprot-TrEMBL)
COX6C(3-75) ProteinP09669 (Uniprot-TrEMBL)
COX7A2L ProteinO14548 (Uniprot-TrEMBL)
COX7B ProteinP24311 (Uniprot-TrEMBL)
COX7C ProteinP15954 (Uniprot-TrEMBL)
COX8A ProteinP10176 (Uniprot-TrEMBL)
CYCS ProteinP99999 (Uniprot-TrEMBL)
CuA MetaboliteCHEBI:28694 (ChEBI)
Cytochrome c (oxidised)ComplexR-HSA-352607 (Reactome)
Cytochrome c (reduced)ComplexR-HSA-352609 (Reactome)
Cytochrome c oxidaseComplexR-HSA-164316 (Reactome)
D-Fructose 2,6-bisphosphateMetaboliteCHEBI:28602 (ChEBI)
D-Glucono-1,5-lactone 6-phosphateMetaboliteCHEBI:16938 (ChEBI)
DDIT4 Gene ProteinENSG00000168209 (Ensembl)
DDIT4 GeneGeneProductENSG00000168209 (Ensembl)
DDIT4 ProteinQ9NX09 (Uniprot-TrEMBL)
DDIT4:14-3-3 dimerComplexR-HSA-5632741 (Reactome)
DDIT4ProteinQ9NX09 (Uniprot-TrEMBL)
Detoxification of

Reactive Oxygen

Species
PathwayR-HSA-3299685 (Reactome) Reactive oxygen species such as superoxide (O2.-), peroxides (ROOR), singlet oxygen, peroxynitrite (ONOO-), and hydroxyl radical (OH.) are generated by cellular processes such as respiration (reviewed in Murphy 2009, Brand 2010) and redox enzymes and are required for signaling yet they are damaging due to their high reactivity (reviewed in Imlay 2008, Buettner 2011, Kavdia 2011, Birben et al. 2012, Ray et al. 2012). Aerobic cells have defenses that detoxify reactive oxygen species by converting them to less reactive products. Superoxide dismutases convert superoxide to hydrogen peroxide and oxygen (reviewed in Fukai and Ushio-Fukai 2011). Catalase and peroxidases then convert hydrogen peroxide to water.
Humans contain 3 superoxide dismutases: SOD1 is located in the cytosol and mitochondrial intermembrane space, SOD2 is located in the mitochondrial matrix, and SOD3 is located in the extracellular region. Superoxide, a negative ion, is unable to easily cross membranes and tends to remain in the compartment where it was produced. Hydrogen peroxide, one of the products of superoxide dismutase, is able to diffuse across membranes and pass through aquaporin channels. In most cells the primary source of hydrogen peroxide is mitochondria and, once in the cytosol, hydrogen peroxide serves as a signaling molecule to regulate redox-sensitive proteins such as transcription factors, kinases, phosphatases, ion channels, and others (reviewed in Veal and Day 2011, Ray et al. 2012). Hydrogen peroxide is decomposed to water by catalase, decomposed to water plus oxidized thioredoxin by peroxiredoxins, and decomposed to water plus oxidized glutathione by glutathione peroxidases (Presnell et al. 2013).
EIF2C1 ProteinQ9UL18 (Uniprot-TrEMBL)
EIF2C3 ProteinQ9H9G7 (Uniprot-TrEMBL)
EIF2C4 ProteinQ9HCK5 (Uniprot-TrEMBL)
Energy dependent

regulation of mTOR

by LKB1-AMPK
PathwayR-HSA-380972 (Reactome) Upon formation of a trimeric LKB1:STRAD:MO25 complex, LKB1 phosphorylates and activates AMPK. This phosphorylation is immediately removed in basal conditions by PP2C, but if the cellular AMP:ATP ratio rises, this activation is maintained, as AMP binding by AMPK inhibits the dephosphorylation. AMPK then activates the TSC complex by phosphorylating TSC2. Active TSC activates the intrinsic GTPase activity of Rheb, resulting in GDP-loaded Rheb and inhibition of mTOR pathway.
FAD MetaboliteCHEBI:16238 (ChEBI)
Fru(6)PMetaboliteCHEBI:57634 (ChEBI)
G6PMetaboliteCHEBI:58225 (ChEBI)
G6PD ProteinP11413 (Uniprot-TrEMBL)
G6PD dimer and tetramerComplexR-HSA-464971 (Reactome)
GDP MetaboliteCHEBI:17552 (ChEBI)
GLS ProteinO94925 (Uniprot-TrEMBL)
GLS dimersComplexR-HSA-507859 (Reactome)
GLS2 Gene ProteinENSG00000135423 (Ensembl)
GLS2 GeneGeneProductENSG00000135423 (Ensembl)
GLS2 ProteinQ9UI32 (Uniprot-TrEMBL)
GLS2 dimerComplexR-HSA-507858 (Reactome)
GPI ProteinP06744 (Uniprot-TrEMBL)
GPI dimerComplexR-HSA-70469 (Reactome)
GPX2 tetramerComplexR-HSA-2142735 (Reactome)
GSHMetaboliteCHEBI:16856 (ChEBI)
GSR-2 ProteinP00390-2 (Uniprot-TrEMBL)
GSR-2:FAD dimerComplexR-HSA-71680 (Reactome)
GSSGMetaboliteCHEBI:17858 (ChEBI)
GTP MetaboliteCHEBI:15996 (ChEBI)
H+MetaboliteCHEBI:15378 (ChEBI)
H2O2MetaboliteCHEBI:16240 (ChEBI)
H2OMetaboliteCHEBI:15377 (ChEBI)
HOOS-C52-PRDX1 ProteinQ06830 (Uniprot-TrEMBL)
HOOS-C52-PRDX1 dimerComplexR-HSA-5631882 (Reactome)
L-GlnMetaboliteCHEBI:58359 (ChEBI)
L-GluMetaboliteCHEBI:29985 (ChEBI)
L-selenocysteine-residue-GPX2 ProteinP18283 (Uniprot-TrEMBL)
LAMTOR1 ProteinQ6IAA8 (Uniprot-TrEMBL)
LAMTOR2 ProteinQ9Y2Q5 (Uniprot-TrEMBL)
LAMTOR3 ProteinQ9UHA4 (Uniprot-TrEMBL)
LAMTOR4 ProteinQ0VGL1 (Uniprot-TrEMBL)
LAMTOR5 ProteinO43504 (Uniprot-TrEMBL)
LRPPRC ProteinP42704 (Uniprot-TrEMBL)
MLST8 ProteinQ9BVC4 (Uniprot-TrEMBL)
MOV10 ProteinQ9HCE1 (Uniprot-TrEMBL)
MT-CO1 ProteinP00395 (Uniprot-TrEMBL)
MT-CO2 ProteinP00403 (Uniprot-TrEMBL)
MT-CO3 ProteinP00414 (Uniprot-TrEMBL)
MTOR ProteinP42345 (Uniprot-TrEMBL)
Metabolism of carbohydratesPathwayR-HSA-71387 (Reactome) Starches and sugars are major constituents of the human diet and the catabolism of monosaccharides, notably glucose, derived from them is an essential part of human energy metabolism (Dashty 2013). Glucose can be catabolized to pyruvate (glycolysis) and pyruvate synthesized from diverse sources can be metabolized to form glucose (gluconeogenesis). Glucose can be polymerized to form glycogen under conditions of glucose excess (glycogen synthesis), and glycogen can be broken down to glucose in response to stress or starvation (glycogenolysis). Other monosaccharides prominent in the diet, fructose and galactose, can be converted to glucose. The disaccharide lactose, the major carbohydrate in breast milk, is synthesized in the lactating mammary gland. The pentose phosphate pathway allows the synthesis of diverse monosaccharides from glucose including the pentose ribose-5-phosphate and the regulatory molecule xylulose-5-phosphate, as well as the generation of reducing equivalents for biosynthetic processes. Glycosaminoglycan metabolism and xylulose-5-phosphate synthesis from glucuronate are also annotated as parts of carbohydrate metabolism.

The digestion of dietary starch and sugars and the uptake of the resulting monosaccharides into the circulation from the small intestine are annotated as parts of the “Digestion and absorption� pathway.

Metabolism of nucleotidesPathwayR-HSA-15869 (Reactome) Nucleotides and their derivatives are used for short-term energy storage (ATP, GTP), for intra- and extra-cellular signaling (cAMP; adenosine), as enzyme cofactors (NAD, FAD), and for the synthesis of DNA and RNA. Most dietary nucleotides are consumed by gut flora; the human body's own supply of these molecules is synthesized de novo. Additional metabolic pathways allow the interconversion of nucleotides, the salvage and reutilization of nucleotides released by degradation of DNA and RNA, the catabolism of excess nucleotides, and the transport of these molecules between the cytosol and the nucleus (Rudolph 1994). These pathways are regulated to control the total size of the intracellular nucleotide pool, to balance the relative amounts of individual nucleotides, and to couple the synthesis of deoxyribonucleotides to the onset of DNA replication (S phase of the cell cycle).

These pathways are also of major clinical interest as they are the means by which nucleotide analogues used as anti-viral and anti-tumor drugs are taken up by cells, activated, and catabolized (Weilin and Nordlund 2010). As well, differences in nucleotide metabolic pathways between humans and aplicomplexan parasites like Plasmodium have been exploited to design drugs to attack the latter (Hyde 2007).

The movement of nucleotides and purine and pyrimidine bases across lipid bilayer membranes, mediated by SLC transporters, is annotated as part of the module "transmembrane transport of small molecules".

Metabolism of amino

acids and

derivatives
PathwayR-HSA-71291 (Reactome) Cellular metabolism of amino acids and related molecules includes the pathways for the catabolism of amino acids, the biosynthesis of the nonessential amino acids (alanine, arginine, aspartate, asparagine, cysteine, glutamate, glutamine, glycine, proline, and serine) and selenocysteine, the synthesis of urea, and the metabolism of carnitine, creatine, choline, polyamides, melanin, and amine-derived hormones. The metabolism of amino acids provides a balanced supply of amino acids for protein synthesis. In the fasting state, the catabolism of amino acids derived from breakdown of skeletal muscle protein and other sources is coupled to the processes of gluconeogenesis and ketogenesis to meet the body’s energy needs in the absence of dietary energy sources. These metabolic processes also provide the nitrogen atoms for the biosynthesis of nucleotides and heme, annotated as separate metabolic processes (Felig 1975; Häussinger 1990; Owen et al. 1979).

Transport of these molecuels across lipid bilayer membranes is annotated separately as part of the module on "transmembrane transport of small molecules".

NADP+ MetaboliteCHEBI:18009 (ChEBI)
NADP+MetaboliteCHEBI:18009 (ChEBI)
NADPHMetaboliteCHEBI:16474 (ChEBI)
NDUFA4 ProteinO00483 (Uniprot-TrEMBL)
NH4+MetaboliteCHEBI:28938 (ChEBI)
O2MetaboliteCHEBI:15379 (ChEBI)
PIP3 activates AKT signalingPathwayR-HSA-1257604 (Reactome) Signaling by AKT is one of the key outcomes of receptor tyrosine kinase (RTK) activation. AKT is activated by the cellular second messenger PIP3, a phospholipid that is generated by PI3K. In ustimulated cells, PI3K class IA enzymes reside in the cytosol as inactive heterodimers composed of p85 regulatory subunit and p110 catalytic subunit. In this complex, p85 stabilizes p110 while inhibiting its catalytic activity. Upon binding of extracellular ligands to RTKs, receptors dimerize and undergo autophosphorylation. The regulatory subunit of PI3K, p85, is recruited to phosphorylated cytosolic RTK domains either directly or indirectly, through adaptor proteins, leading to a conformational change in the PI3K IA heterodimer that relieves inhibition of the p110 catalytic subunit. Activated PI3K IA phosphorylates PIP2, converting it to PIP3; this reaction is negatively regulated by PTEN phosphatase. PIP3 recruits AKT to the plasma membrane, allowing TORC2 to phosphorylate a conserved serine residue of AKT. Phosphorylation of this serine induces a conformation change in AKT, exposing a conserved threonine residue that is then phosphorylated by PDPK1 (PDK1). Phosphorylation of both the threonine and the serine residue is required to fully activate AKT. The active AKT then dissociates from PIP3 and phosphorylates a number of cytosolic and nuclear proteins that play important roles in cell survival and metabolism. For a recent review of AKT signaling, please refer to Manning and Cantley, 2007.
PRDX1 ProteinQ06830 (Uniprot-TrEMBL)
PRDX1 dimerComplexR-HSA-3341319 (Reactome)
PRDX1,2,5ComplexR-HSA-3341359 (Reactome)
PRDX2 ProteinP32119 (Uniprot-TrEMBL)
PRDX5 ProteinP30044-2 (Uniprot-TrEMBL)
PRKAB1 ProteinQ9Y478 (Uniprot-TrEMBL)
PRKAB2 ProteinO43741 (Uniprot-TrEMBL)
PRKAG1 ProteinP54619 (Uniprot-TrEMBL)
PRKAG2 ProteinQ9UGJ0 (Uniprot-TrEMBL)
PRKAG3 ProteinQ9UGI9 (Uniprot-TrEMBL)
PTEN gene ProteinENSG00000171862 (Ensembl)
PTEN geneGeneProductENSG00000171862 (Ensembl)
PTEN mRNA ProteinENST00000371953 (Ensembl)
PTEN mRNA:miR-26A RISCComplexR-HSA-2318750 (Reactome)
PTEN mRNARnaENST00000371953 (Ensembl)
PTENProteinP60484 (Uniprot-TrEMBL)
PiMetaboliteCHEBI:43474 (ChEBI)
RHEB ProteinQ15382 (Uniprot-TrEMBL)
RPTOR ProteinQ8N122 (Uniprot-TrEMBL)
RRAGA ProteinQ7L523 (Uniprot-TrEMBL)
RRAGB ProteinQ5VZM2 (Uniprot-TrEMBL)
RRAGC ProteinQ9HB90 (Uniprot-TrEMBL)
RRAGD ProteinQ9NQL2 (Uniprot-TrEMBL)
RRM2B Gene ProteinENSG00000048392 (Ensembl)
RRM2B GeneGeneProductENSG00000048392 (Ensembl)
RRM2BProteinQ7LG56 (Uniprot-TrEMBL)
Respiratory electron

transport, ATP synthesis by chemiosmotic coupling, and heat production by uncoupling

proteins.
PathwayR-HSA-163200 (Reactome) Oxidation of fatty acids and pyruvate in the mitochondrial matrix yield large amounts of NADH. The respiratory electron transport chain couples the re-oxidation of this NADH to NAD+ to the export of protons from the mitochonrial matrix, generating a chemiosmotic gradient across the inner mitochondrial membrane. This gradient is used to drive the synthesis of ATP; it can also be bypassed by uncoupling proteins to generate heat, a reaction in brown fat that may be important in regulation of body temperature in newborn children.
SCO1 ProteinO75880 (Uniprot-TrEMBL)
SCO2 Gene ProteinENSG00000130489 (Ensembl)
SCO2 GeneGeneProductENSG00000130489 (Ensembl)
SCO2 ProteinO43819 (Uniprot-TrEMBL)
SCO2ProteinO43819 (Uniprot-TrEMBL)
SESN1 Gene ProteinENSG00000080546 (Ensembl)
SESN1,2,3 GenesComplexR-HSA-5629150 (Reactome)
SESN1,2,3:HOOS-C52-PRDX1 dimerComplexR-HSA-5631902 (Reactome)
SESN1,2,3:p-AMPK heterotrimer:AMPComplexR-HSA-5631939 (Reactome)
SESN1,2,3ComplexR-HSA-5629191 (Reactome)
SESN1-1,SESN1-3 R-HSA-5629186 (Reactome)
SESN2 Gene ProteinENSG00000130766 (Ensembl)
SESN2 ProteinP58004 (Uniprot-TrEMBL)
SESN3 Gene ProteinENSG00000149212 (Ensembl)
SESN3 ProteinP58005 (Uniprot-TrEMBL)
SFN ProteinP31947 (Uniprot-TrEMBL)
SLC38A9 ProteinQ8NBW4 (Uniprot-TrEMBL)
SURF1 ProteinQ15526 (Uniprot-TrEMBL)
TACO1 ProteinQ9BSH4 (Uniprot-TrEMBL)
TIGAR Gene ProteinENSG00000078237 (Ensembl)
TIGAR GeneGeneProductENSG00000078237 (Ensembl)
TIGARProteinQ9NQ88 (Uniprot-TrEMBL)
TNRC6A ProteinQ8NDV7 (Uniprot-TrEMBL)
TNRC6B ProteinQ9UPQ9 (Uniprot-TrEMBL)
TNRC6C ProteinQ9HCJ0 (Uniprot-TrEMBL)
TNXRD1:FAD dimerComplexR-HSA-73532 (Reactome)
TP53

Tetramer:SESN1,2,3

Genes
ComplexR-HSA-5629180 (Reactome)
TP53 ProteinP04637 (Uniprot-TrEMBL)
TP53 Tetramer:GLS2 GeneComplexR-HSA-5632919 (Reactome)
TP53 Tetramer:PTEN GeneComplexR-HSA-5632941 (Reactome)
TP53 Tetramer:RRM2B GeneComplexR-HSA-5632886 (Reactome)
TP53 Tetramer:SCO2 GeneComplexR-HSA-5632755 (Reactome)
TP53 Tetramer:TIGAR GeneComplexR-HSA-5628900 (Reactome)
TP53 TetramerComplexR-HSA-3209194 (Reactome)
TP63 ProteinQ9H3D4 (Uniprot-TrEMBL)
TP63 Tetramer/ TP53 TetramerComplexR-HSA-5632387 (Reactome)
TP63/T53:DDIT4 GeneComplexR-HSA-5632392 (Reactome)
TSC1 ProteinQ92574 (Uniprot-TrEMBL)
TSC1:TSC2ComplexR-HSA-165175 (Reactome)
TSC1:p-S1387-TSC2ComplexR-HSA-381855 (Reactome)
TSC1ProteinQ92574 (Uniprot-TrEMBL)
TSC2 ProteinP49815 (Uniprot-TrEMBL)
TSC2ProteinP49815 (Uniprot-TrEMBL)
TXNProteinP10599 (Uniprot-TrEMBL)
TXNRD1 ProteinQ16881 (Uniprot-TrEMBL)
YWHAB ProteinP31946 (Uniprot-TrEMBL)
YWHAE ProteinP62258 (Uniprot-TrEMBL)
YWHAG ProteinP61981 (Uniprot-TrEMBL)
YWHAH ProteinQ04917 (Uniprot-TrEMBL)
YWHAQ ProteinP27348 (Uniprot-TrEMBL)
YWHAZ ProteinP63104 (Uniprot-TrEMBL)
ferriheme MetaboliteCHEBI:38574 (ChEBI)
ferroheme MetaboliteCHEBI:38573 (ChEBI)
mTORC1:Ragulator:Rag:GNP:RHEB:GDPComplexR-HSA-5693447 (Reactome)
miR-26A RISCComplexR-HSA-2318737 (Reactome)
miR-26A1 ProteinMI0000083 (miRBase mature sequence)
miR-26A2 ProteinMI0000750 (miRBase mature sequence)
p-AMPK heterotrimer:AMPComplexR-HSA-380931 (Reactome)
p-S1387-TSC2 ProteinP49815 (Uniprot-TrEMBL)
p-S939,T1462-TSC2 ProteinP49815 (Uniprot-TrEMBL)
p-S939,T1462-TSC2:14-3-3 dimerComplexR-HSA-5632727 (Reactome)
p-S939,T1462-TSC2ProteinP49815 (Uniprot-TrEMBL)
p-T,p-S-AKTComplexR-HSA-202074 (Reactome)
p-T172-PRKAA2 ProteinP54646 (Uniprot-TrEMBL)
p-T183-PRKAA1 ProteinQ13131 (Uniprot-TrEMBL)
p-T305,S472-AKT3 ProteinQ9Y243 (Uniprot-TrEMBL)
p-T308,S473-AKT1 ProteinP31749 (Uniprot-TrEMBL)
p-T309,S474-AKT2 ProteinP31751 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
14-3-3 dimerR-HSA-5632732 (Reactome)
14-3-3 dimerR-HSA-5632738 (Reactome)
2xHC-TXNArrowR-HSA-3341343 (Reactome)
2xHC-TXNR-HSA-73646 (Reactome)
ADPArrowR-HSA-198609 (Reactome)
ADPArrowR-HSA-380927 (Reactome)
ATPR-HSA-198609 (Reactome)
ATPR-HSA-380927 (Reactome)
Active mTORC1 complexR-HSA-380979 (Reactome)
Active mTORC1 complexmim-catalysisR-HSA-380979 (Reactome)
COX ancilliary proteinsArrowR-HSA-163214 (Reactome)
COX11,14,16,18,20ArrowR-HSA-163214 (Reactome)
COX19ArrowR-HSA-163214 (Reactome)
Cytochrome c (oxidised)ArrowR-HSA-163214 (Reactome)
Cytochrome c (reduced)R-HSA-163214 (Reactome)
Cytochrome c oxidasemim-catalysisR-HSA-163214 (Reactome)
D-Fructose 2,6-bisphosphateR-HSA-5628905 (Reactome)
D-Glucono-1,5-lactone 6-phosphateArrowR-HSA-70377 (Reactome)
DDIT4 GeneR-HSA-5632386 (Reactome)
DDIT4 GeneR-HSA-5632393 (Reactome)
DDIT4:14-3-3 dimerArrowR-HSA-5632738 (Reactome)
DDIT4:14-3-3 dimerTBarR-HSA-5632732 (Reactome)
DDIT4ArrowR-HSA-5632386 (Reactome)
DDIT4R-HSA-5632738 (Reactome)
Fru(6)PArrowR-HSA-5628905 (Reactome)
Fru(6)PR-HSA-70475 (Reactome)
G6PArrowR-HSA-70475 (Reactome)
G6PD dimer and tetramermim-catalysisR-HSA-70377 (Reactome)
G6PR-HSA-70377 (Reactome)
GLS dimersmim-catalysisR-HSA-70609 (Reactome)
GLS2 GeneR-HSA-5632914 (Reactome)
GLS2 GeneR-HSA-5632924 (Reactome)
GLS2 dimerArrowR-HSA-5632924 (Reactome)
GPI dimermim-catalysisR-HSA-70475 (Reactome)
GPX2 tetramermim-catalysisR-HSA-3341277 (Reactome)
GSHArrowR-HSA-71682 (Reactome)
GSHR-HSA-3341277 (Reactome)
GSR-2:FAD dimermim-catalysisR-HSA-71682 (Reactome)
GSSGArrowR-HSA-3341277 (Reactome)
GSSGR-HSA-71682 (Reactome)
H+ArrowR-HSA-163214 (Reactome)
H+ArrowR-HSA-70377 (Reactome)
H+R-HSA-163214 (Reactome)
H+R-HSA-71682 (Reactome)
H+R-HSA-73646 (Reactome)
H2O2R-HSA-3341277 (Reactome)
H2O2R-HSA-3341343 (Reactome)
H2O2R-HSA-5631885 (Reactome)
H2OArrowR-HSA-163214 (Reactome)
H2OArrowR-HSA-3341277 (Reactome)
H2OArrowR-HSA-3341343 (Reactome)
H2OArrowR-HSA-5631885 (Reactome)
H2OR-HSA-5628905 (Reactome)
H2OR-HSA-70609 (Reactome)
HOOS-C52-PRDX1 dimerArrowR-HSA-5631885 (Reactome)
HOOS-C52-PRDX1 dimerR-HSA-5631903 (Reactome)
L-GlnR-HSA-70609 (Reactome)
L-GluArrowR-HSA-70609 (Reactome)
NADP+ArrowR-HSA-71682 (Reactome)
NADP+ArrowR-HSA-73646 (Reactome)
NADP+R-HSA-70377 (Reactome)
NADPHArrowR-HSA-70377 (Reactome)
NADPHR-HSA-71682 (Reactome)
NADPHR-HSA-73646 (Reactome)
NH4+ArrowR-HSA-70609 (Reactome)
O2R-HSA-163214 (Reactome)
PRDX1 dimerR-HSA-5631885 (Reactome)
PRDX1 dimermim-catalysisR-HSA-5631885 (Reactome)
PRDX1,2,5mim-catalysisR-HSA-3341343 (Reactome)
PTEN geneR-HSA-5632939 (Reactome)
PTEN geneR-HSA-5632993 (Reactome)
PTEN mRNA:miR-26A RISCArrowR-HSA-2318752 (Reactome)
PTEN mRNA:miR-26A RISCTBarR-HSA-2321904 (Reactome)
PTEN mRNAArrowR-HSA-5632993 (Reactome)
PTEN mRNAR-HSA-2318752 (Reactome)
PTEN mRNAR-HSA-2321904 (Reactome)
PTENArrowR-HSA-2321904 (Reactome)
PiArrowR-HSA-380979 (Reactome)
PiArrowR-HSA-5628905 (Reactome)
R-HSA-163214 (Reactome) Complex IV (COX, cytochrome c oxidase) contains the hemeprotein cytochrome a and a3. It also contains copper atoms which undergo a transition from Cu+ to Cu2+ during the transfer of electrons through the complex to molecular oxygen. A bimetallic centre containing a copper atom and a heme-linked iron protein binds oxygen after 4 electrons have been picked up. Water, the final product of oxygen reduction, is then released. Oxygen is the final electron acceptor in the respiratory chain. The overall reaction can be summed as

4Cyt c (red.) + 12H+ (in) + O2 = 4Cyt c (ox.) + 2H2O + 8H+ (out)

Four protons are taken up from the matrix side of the membrane to form the water (scalar protons). Wikstrom (1977) suggests 4 protons are additionally transferred out from the matrix to the intermembrane space.

COX ancillary proteins mediate membrane insertion, catalytic core processing, copper transport and insertion into core subunits and heme A biosynthesis (Stilburek et al. 2006, Fontanesi et al. 2006, Soto et al. 2012). To date, all Mendelian disorders presenting COX deficiency have been assigned to mutations in ancillary factors, with the exception of an infantile encephalomyopathy caused by a defective COX6B1 and an exocrine pancreatic insufficiency caused by a defective COX4I2 gene (Soto et al. 2012). Balsa et al have shown that NDUFA4, formerly considered to be a constituent of NADH dehydrogenase (Complex I), is instead a component of the cytochrome c oxidase (CIV) (Balsa et al. 2012). Patients with NDUFA4 mutations display COX deficiencies (Pitceathly et al. 2013).
R-HSA-165179 (Reactome) A membrane-associated TSC1 (hamartin) binds TSC2 (tuberin) and recruits it to the plasma membrane where it can exert its function as a GAP (GTPase activating protein) for the small GTPase RHEB (Cai et al. 2006).
R-HSA-198609 (Reactome) AKT phosphorylates and inhibits TSC2 (tuberin), a suppressor of the TOR kinase pathway, which senses nutrient levels in the environment. TSC2 forms a protein complex with TSC1 and this complex acts as a GAP (GTPase activating protein) for the RHEB G-protein. RHEB, in turn, activates the TOR kinase. Thus, an active AKT1 activates the TOR kinase, both of which are positive signals for cell growth (an increase in cell mass) and division.
The TOR kinase regulates two major processes: translation of selected mRNAs in the cell and autophagy. In the presence of high nutrient levels TOR is active and phosphorylates the 4EBP protein releasing the eukaryotic initiation factor 4E (eIF4E), which is essential for cap-dependent initiation of translation and promoting growth of the cell (PMID: 15314020). TOR also phosphorylates the S6 kinase, which is implicated in ribosome biogenesis as well as in the modification of the S6 ribosomal protein. AKT can also activate mTOR by another mechanism, involving phosphorylation of PRAS40, an inhibitor of mTOR activity.
R-HSA-2318752 (Reactome) MIR26A microRNAs, miR-26A1 and miR-26A2, transcribed from genes on chromosome 3 and 12, respectively, bind PTEN mRNA (Huse et al. 2009).

The MIR26A2 locus is frequently amplified in glioma tumors that retain one wild-type PTEN allele. The resulting miR-26A2 overexpression leads to down-regulation of PTEN protein level. Overexpression of miR-26A2 was shown to enhance tumorigenesis and negatively correlates with the loss of heterozygosity at the PTEN locus in a mouse PTEN +/- glioma model, based on monoallelic PTEN loss (Huse et al. 2009, Kim et al. 2010).
R-HSA-2321904 (Reactome) PTEN protein synthesis is negatively regulated by microRNAs miR-26A1 and miR-26A2, which recruit the RISC complex to PTEN mRNA. Overexpression of miR-26A2, caused by genomic amplification of MIR26A2 locus on chromosome 12, is frequently observed in human brain glioma tumors possessing one wild-type PTEN allele, and is thought to contribute to tumor progression by repressing PTEN protein expression from the remaining allele (Huse et al. 2009). Other microRNAs, which may also be altered in cancer, such as miR-17, miR-19a, miR-19b, miR-20a, miR-20b, miR-21, miR-22, miR-25, miR-93, miR-106a, miR-106b, miR 205, and miR 214, also bind PTEN mRNA and inhibit its translation into protein (Meng et al. 2007, Xiao et al. 2008, Yang et al. 2008, Kim et al. 2010, Poliseno, Salmena, Riccardi et al. 2010, Zhang et al. 2010, Tay et al. 2011, Qu et al. 2012, Cai et al. 2013).
R-HSA-3341277 (Reactome) GPX2 (located in the gastrointestinal tract, also called GSHPx-GI, GPX-GI, and GI-GPx), like glutathione peroxidase 1 (GPX1, ubiquitous), reduces one molecule of hydrogen peroxide (H2O2) with two molecules of glutathione to yield one molecule of oxidized glutathione (glutathione disulfide, GSSG) and two molecules of water (Chu et al. 1998).
R-HSA-3341343 (Reactome) Peroxiredoxin 1 (PRDX1), PRDX2, and PRDX5 in the cytosol reduce hydrogen peroxide (H2O2) with thioredoxin yielding oxidized thioredoxin and water (Yamashita et al. 1999, Lee et al. 2007, Nagy et al. 2011).
R-HSA-380927 (Reactome) Activated AMPK (phosphorylated on the alpha subunit and with AMP bound) phosphorylates TSC2 (also known as tuberin) on Ser-1387, thereby activating the GTPase activating protein (GAP) activity of the Tuberous Sclerosis Complex (TSC). The TSC tumor suppressor is a critical upstream inhibitor of the mTORC1 complex. TSC is a GTPase-activating protein that stimulates the intrinsic GTPase activity of the small G-protein Rheb. This inactivates Rheb by stimulating its GTPase activity. The GDP-bound form of Rheb looses the ability to activate the kinase activity of the mTORC1 complex (Sancak et al. 2007). Loss of TSC1 or TSC2 leads to hyperactivation of mTORC1.

Phosphorylation of TSC1 and TSC2 serves as an integration point for a wide variety of environmental signals that regulate mTORC1 (Sabatini 2006). Mitogen-activated kinases including Akt, Erk, and Rsk directly phosphorylate TSC2, leading to its inactivation by an unknown mechanism. Another Akt substrate, PRAS40, was recently shown to bind and inhibit the mTORC1 complex. Upon phosphorylation by Akt, PRAS40 no longer inhibits mTORC1 (Sancak et al. 2007; Vander Haar et al. 2007).
R-HSA-380979 (Reactome) TSC2 (in the TSC complex) functions as a GTPase-activating protein and stimulates the intrinsic GTPase activity of the small G-protein Rheb. This results in the conversion of Rheb:GTP to Rheb:GDP. GDP-bound Rheb is unable to activate mTOR (Inoki et al. 2003, Tee et al. 2003). It is not demonstrated that RHEB hydrolyzes GTP when present in the mTORC1 complex; given the low affinity of RHEB for mTOR, it may dissociate from the mTORC1 complex before TSC2 stimulates hydrolysis of GTP; TSC2 may not have access to critical residues of RHEB when present inside mTORC1.
R-HSA-5628899 (Reactome) TIGAR gene possesses two TP53 (p53) binding sites, one upstream of the first exon and another within the first intron. TP53 can bind both sites, with a higher affinity for the intronic site (Bensaad et al. 2006).
R-HSA-5628901 (Reactome) TIGAR was first identified as a TP53 target through high-throughput gene expression profiling (Jen and Cheung 2005). TP53 stimulates TIGAR transcription, although TIGAR can be regulated through TP53-independent mechanisms, including TP53 family members TP63 (p63) and TP73 (p73). TIGAR is induced by TP53 under low stress levels and decreases under high stress levels (Bensaad et al. 2006). TIGAR functions as a fructose-2,6-bisphosphatase, thereby lowering glycolytic flux and promoting antioxidant functions. By protecting cells from oxidative stress, TIGAR may mediate some of the tumor suppressor activity of p53 but could also contribute to tumorigenesis. (Bensaad, 2006, Lee et al. 2014).
R-HSA-5628905 (Reactome) TIGAR shares similarity with PGMs (phosphoglycerate mutases), especially PFK2 (6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase). TIGAR possesses only the bisphosphatase domain and converts D-fructose 2,6-bisphosphate into D-fructose 6-phosphate (Bensaad et al. 2006). Reduction of fructose 2,6-bisphosphate levels correlates with decrease in glycolytic rates, which makes cells more sensitive to apoptotic stimuli (Vander Heiden et al. 2001). Alternatively, fructose 6-phosphate can be isomerized to glucose 6-phosphate, which is diverted to the pentose phosphate pathway, which can have an anti-apoptotic effect (Boada et al. 2000, Perez et al. 2000). In the pentose phosphate pathway, oxidized glutathione is reduced, and this reduced glutathione can then be used by glutathione peroxidase to remove hydrogen peroxide, thereby protecting cells from the oxidative stress (Kletzien et al. 1994, Fico et al. 2004, Tian et al. 1999). Indeed, expression of TIGAR increases reduced glutathione to oxidized glutathione ratio and lowers ROS (reactive oxygen species) levels in cells (Bensaad et al. 2006, Lee et al. 2014).
R-HSA-5629187 (Reactome) TP53 (p53) binds to the p53 response element in the intron 2 of SESN1 gene and stimulates transcription of SESN1 transcripts SESN1-1 and SESN1-3, also known as PA26 T2 and PA26 T3 (Velasco-Miguel et al. 1999). Recently, TP53 binding to SESN2 gene regulatory elements has been identified by ChIPseq (Menendez et al. 2013), and SESN2 gene expression was previously shown to be responsive to TP53 (Budanov et al. 2002). Rat ortholog of SESN3 was shown to possess p53 binding sites in the promoter region, but direct binding of TP53 to regulatory elements of human SESN3 has not been examined (Brynczka et al. 2007).
R-HSA-5629189 (Reactome) Sestrins (SESN) are a small family of stress-sensitive gene that are conserved across several species. Mammals express three different SESN family members characterized as SESN1-3. Sestrin genes, SESN1, SESN2 and SESN3, are upregulated in response to TP53-mediated transcriptional regulation. SESN1 and SESN2 were classified as members of the growth arrest and DNA damage (GADD) gene family that can regulate cell growth and viability under different cellular pressures. In particular, p53 negatively modulates the mTOR pahtway via SESN1 and SESN2 upregulation (Feng 2010). SESN3 was identified shortly after SESN2 through in silico analysis and was found to be a target of the forkhead transcription factor (FOXO) family. A specific TP53 binding site on the human SESN3 promoter has not been identified yet, but was found in the rat ortholog (Velasco-Miguel et al. 1999, Budanov et al. 2002, Brynczka et al. 2007).
R-HSA-5631885 (Reactome) The activity of eukaryotic PRDX1 gradually decreases with time, which is due to the overoxidation of the catalytic cysteine C52. Normally, oxidized cysteine C52-SOH is generated as a catalytic intermediate, which is subsequently reduced by thioredoxin. Occasionally, further oxidation happens, generating C52-SOOH , where the catalytic cysteine is converted to cysteine-sulfinic acid. This over-oxidation cannot be reversed by thioredoxin (Yang et al. 2002, Budanov et al. 2004). Bacterial peroxiredoxin AhpC does not undergo over-oxidation due to structural difference (Wood et al. 2003).
R-HSA-5631903 (Reactome) Sestrins (SESN1, SESN2 and likely SESN3) bind overoxidized PRDX1, in which the catalytic cysteine C52 has been converted to cysteine-sulfinic acid. Among all peroxiredoxins, PRDX1 is the most abundant member of the PRDX family. The major function is to protect cells against reactive oxygen species (ROS), thus impacting on cell proliferation and survival (Gong et al. 2015). While several reports state that sestrins reduce overoxidized PRDX1 to the catalytically active homodimer (Budanov et al. 2004, Papadia et al. 2008, Essler et al. 2009), there are conflicting reports claiming that sestrins do not possess cysteine sulfinyl reductase activity (Woo et al. 2009).
R-HSA-5631941 (Reactome) SESN1, SESN2 and possibly SESN3 are able to bind the AMPK complex and increase its catalytic activity. The exact mechanism has not been elucidated, but recent studies suggest that sestrin-bound AMPK is resistant to inactivation through AKT-induced dephosphorylation (Budanov and Karin 2008, Sanli et al. 2012, Cam et al. 2014).
R-HSA-5632386 (Reactome) Transcription of DDIT4 (REDD1) gene is stimulated by TP63, both during mouse embryonal development and under conditions of genotoxic and oxidative stress. TP53 stimulates DDIT4 transcription after TP53 activation by ionizing radiation, but it seems that TP63 is the main activator of DDIT4 transcription under stress conditions (Ellisen et al. 2002).
R-HSA-5632393 (Reactome) DDIT4 (REDD1) gene has a p53 response element immediately upstream of the transcription start site, and this p53 response element is able to bind both TP63 and TP53 transcription factors (Ellisen et al. 2002).
R-HSA-5632732 (Reactome) Phosphorylation of TSC2 by AKT enables association of TSC2 with 14-3-3 proteins YWHAB (14-3-3 protein beta/alpha), YWHAQ (14-3-3 protein theta), YWHAG (14-3-3 protein gamma), YWHAH (14-3-3 protein eta), YWHAE (14-3-3 protein epsilon), YWHAZ (14-3-3 protein zeta/delta) or SFN (14-3-3 protein sigma) (Liu et al. 2002). Binding to 14-3-3 proteins sequesters TSC2 to the cytosol and prevents its association with TSC1 (Cai et al. 2006).
R-HSA-5632738 (Reactome) DDIT4 (REDD1) binds 14-3-3 proteins through a conserved 14-3-3 binding motif Arg-X-X-X-Ser/Thr-X-Pro (DeYoung et al. 2008). Binding of DDIT4 to 14-3-3 proteins competes with 14-3-3 binding to TSC2 and thus prevents AKT-mediated inactivation of TSC2 (Cam et al. 2014).
R-HSA-5632759 (Reactome) TP53 (p53) binds the p53 response element in the intron 1 of SCO2 (Synthesis of Cytochrome c Oxidase 2) gene (Matoba et al. 2006). The binding of TP53 on SCO2 gene was verified in a genome wide chromatin immunoprecipitation study (Wei et al. 2006). Tp53 was also found to bind to the promoter region in mouse Sco2 gene to stimulate its expression in response to physical exercise (Qi et al. 2011).
R-HSA-5632766 (Reactome) TP53 (p53) directly stimulates transcription of the SCO2 gene. SCO2, synthesis of cytochrome c oxidase 2, is a copper-binding assembly protein for the mitochondrial COX (cytochrome C oxidase) complex which enables aerobic respiration. When SCO2 levels are reduced, as occurs in TP53 deficient cells, the glycolysis becomes the main energy source for the cell. The TP53-mediated regulation of SCO2 and other mitochondrial biogenesis genes provides a possible explanation for the Warburg effect (Warburg 1956) observed in some cancer cells (Matoba et al. 2006).
R-HSA-5632887 (Reactome) TP53 (p53) binds the p53-binding site in the first intron of RRM2B (p53R2) gene, which encodes a subunit of the ribonucleotide reductase complex (Tanaka et al. 2000). RRM2B is also regulated by TP73 (p73), a p53 family member (Nakano et al. 2000).
R-HSA-5632892 (Reactome) TP53 (p53) directly stimulates transcription of RRM2B gene (p53R2), which encodes a critical subunit of the ribonucleotide reductase complex (Tanaka et al. 2000), responsible for de novo conversion of ribonucleotides (NTPs) to deoxyribonucleotides (dNTPs). This regulation provides a direct mechanism through which TP53 contributes to DNA synthesis/repair. Mutations in RRM2B gene cause severe mitochondrial DNA depletion (Bourdon et al. 2007, Kulawiec et al. 2009).
R-HSA-5632914 (Reactome) The mitochondrial glutaminase GLS2 gene possesses two putative p53-binding sites in its promoter and one putative p53 binding site in the first intron. TP53 was demonstrated to bind to p53-response elements in the promoter but not intron 1 of GLS2 (Hu et al. 2010, Suzuki et al. 2010).
R-HSA-5632924 (Reactome) TP53 (p53) directly stimulates transcription of mitochondrial glutaminase GLS2 under non-stress and stress conditions. Increased GLS2 levels lead to increased production of glutamate and alpha-ketoglutarate, increased mitochondrial respiration rate, and reduced ROS (reactive oxygen species) load through enhanced glutathione reduction (Hu et al. 2010).

Elevated GLS2 was associated with lower levels of intracellular ROS and a decrease in DNA oxidation. GLS2 knockdown resulted in higher ROS levels and was associated with stimulation of p53-induced cell death (Suzuki et al. 2010).

R-HSA-5632939 (Reactome) PTEN (phosphatase and tensin homolog deleted in chromosome 10) is a tumor suppressor gene that is deleted or mutated in a variety of human cancers. TP53 (p53) binds to the p53-binding site at the PTEN promoter level (Stambolic et al. 2001).
R-HSA-5632993 (Reactome) PTEN (phosphatase and tensin homolog deleted in chromosome 10) is a tumor suppressor gene that is deleted or mutated in a variety of human cancers. TP53 (p53) stimulates PTEN transcription (Stambolic et al. 2000, Singh et al. 2002). PTEN, acting as a negative regulator of PI3K/AKT signaling, affects cell survival, cell cycling, proliferation and migration. PTEN regulates TP53 stability by inhibiting AKT-mediated activation of TP53 ubiquitin ligase MDM2, and thus enhances TP53 transcriptional activity and its own transcriptional activation by TP53. Beside their cross-regulation, PTEN and TP53 can interact and cooperate to regulate survival or apoptotic phenomena (Stambolic et al. 2000, Singh et al. 2002, Nakanishi et al. 2014).
In response to UV induced DNA damage, PTEN transcription is stimulated by binding of the transcription factor EGR1 to the promoter region of PTEN (Virolle et al. 2001).
PTEN transcription is also stimulated by binding of the activated nuclear receptor PPARG (PPARgamma) to peroxisome proliferator response elements (PPREs) in the promoter of the PTEN gene (Patel et al. 2001), binding of the ATF2 transcription factor, activated by stress kinases of the p38 MAPK family, to ATF response elements in the PTEN gene promoter (Shen et al. 2006) and by the transcription factor MAF1 (Li et al. 2016).
NR2E1 (TLX) associated with transcription repressors binds the evolutionarily conserved TLX consensus site in the PTEN promoter. NR2E1 inhibits PTEN transcription by associating with various transcriptional repressors, probably in a cell type or tissue specific manner. PTEN transcription is inhibited when NR2E1 forms a complex with ATN1 (atrophin-1) (Zhang et al. 2006, Yokoyama et al. 2008), KDM1A (LSD1) histone methyltransferase containing CoREST complex (Yokoyama et al. 2008), or histone deacetylases HDAC3, HDAC5 or HDAC7 (Sun et al. 2007).
Binding of the transcriptional repressor SNAI1 (Snail1) to the PTEN promoter represses PTEN transcription. SNAI1-mediated repression of PTEN transcription may require phosphorylation of SNAI1 on serine residue S246. Binding of SNAI1 to the PTEN promoter increases in response to ionizing radiation and is implicated in SNAI1-mediated resistance to gamma-radiation induced apoptosis (Escriva et al. 2008). Binding of another Slug/Snail family member SNAI2 (SLUG) to the PTEN gene promoter also represses PTEN transcription (Uygur et al. 2015).
Binding of JUN to the AP-1 element in the PTEN gene promoter (Hettinger et al. 2007) inhibits PTEN transcription. JUN partner FOS is not needed for JUN-mediated downregulation of PTEN (Vasudevan et al. 2007).
Binding of the transcription factor SALL4 to the PTEN gene promoter (Yang et al. 2008) and SALL4-medaited recruitment of the transcriptional repressor complex NuRD (Lu et al. 2009, Gao et al. 2013), containing histone deacetylases HDAC1 and HDAC2, inhibits the PTEN gene transcription. SALL4-mediated recruitment of DNA methyltransferases (DNMTs) is also implicated in transcriptional repression of PTEN (Yang et al. 2012).
Binding of the transcription factor MECOM (EVI1) to the PTEN gene promoter and MECOM-mediated recruitment of polycomb repressor complexes containing BMI1 (supposedly PRC1.4), and EZH2 (PRC2) leads to repression of PTEN transcription (Song et al. 2009, Yoshimi et al. 2011).
R-HSA-70377 (Reactome) Cytosolic glucose-6-phosphate dehydrogenase (G6PD) catalyzes the reaction of glucose 6-phosphate and NADP+ to form D-glucono-1,5-lactone 6-phosphate and NADPH + H+. This constitutes the first committed step of the pentose phosphate pathway and it is critical to the maintenance of NAPDH pool and redox homeostasis. For this reason, anti-cancer therapies are making this step as a prominent target in cancer therapy (Zhang et al. 2014). The reaction is inhibited by high ADP/AMP concentration, and by high NAPDH concentration. Biochemical studies indicate that both G6PD dimers and tetramers are catalytically active and present under physiological conditions in vivo (Au et al. 2000). Mutations that reduce the catalytic efficiency of G6PD are remarkably common in human populations; these appear to have a protective effect against malaria (e.g., Luzzatto and Afolayan 1968).
R-HSA-70475 (Reactome) The reversible isomerization of fructose-6-phosphate to form glucose-6-phosphate is catalyzed by cytosolic phosphoglucose isomerase (Noltman 1972; Xu and Beutler 1994; Tsuboi et al. 1958).
R-HSA-70609 (Reactome) Mitochondrial glutaminase (GLS) catalyzes the hydrolysis of glutamine to yield glutamate and ammonia. Two GLS enzymes have been identified, one abundantly expressed in the liver (GLS - Elgadi et al. 1999) and one abundantly expressed in kidney (GLS2 - Gomez-Fabre et al. 2000). Their biochemical properties are similar. The enzymes are inferred to function as dimers based on unpublished crystallographic data for GLS (PDB 3CZD) and studies of glutaminase enzyme purified from Ehrlich Ascites cells (Quesada et al. 1988).
R-HSA-71682 (Reactome) Cytosolic glutathione reductase catalyzes the reaction of glutathione (oxidized) and NADPH + H+ to form two molecules of glutathione (reduced) and NADP+ (Scott et al. 1963, Loos et al. 1976). Deficiency of glutathione reductase can cause hemolytic anemia.
R-HSA-73646 (Reactome) Cytosolic thioredoxin reductase catalyzes the reaction of thioredoxin, oxidized and NADPH + H+ to form thioredoxin, reduced and NADP+ (Urig et al. 2006).
RRM2B GeneR-HSA-5632887 (Reactome)
RRM2B GeneR-HSA-5632892 (Reactome)
RRM2BArrowR-HSA-5632892 (Reactome)
SCO2 GeneR-HSA-5632759 (Reactome)
SCO2 GeneR-HSA-5632766 (Reactome)
SCO2ArrowR-HSA-5632766 (Reactome)
SESN1,2,3 GenesR-HSA-5629187 (Reactome)
SESN1,2,3 GenesR-HSA-5629189 (Reactome)
SESN1,2,3:HOOS-C52-PRDX1 dimerArrowR-HSA-5631903 (Reactome)
SESN1,2,3:p-AMPK heterotrimer:AMPArrowR-HSA-5631941 (Reactome)
SESN1,2,3ArrowR-HSA-5629189 (Reactome)
SESN1,2,3R-HSA-5631903 (Reactome)
SESN1,2,3R-HSA-5631941 (Reactome)
TIGAR GeneR-HSA-5628899 (Reactome)
TIGAR GeneR-HSA-5628901 (Reactome)
TIGARArrowR-HSA-5628901 (Reactome)
TIGARmim-catalysisR-HSA-5628905 (Reactome)
TNXRD1:FAD dimermim-catalysisR-HSA-73646 (Reactome)
TP53

Tetramer:SESN1,2,3

Genes
ArrowR-HSA-5629187 (Reactome)
TP53

Tetramer:SESN1,2,3

Genes
ArrowR-HSA-5629189 (Reactome)
TP53 Tetramer:GLS2 GeneArrowR-HSA-5632914 (Reactome)
TP53 Tetramer:GLS2 GeneArrowR-HSA-5632924 (Reactome)
TP53 Tetramer:PTEN GeneArrowR-HSA-5632939 (Reactome)
TP53 Tetramer:PTEN GeneArrowR-HSA-5632993 (Reactome)
TP53 Tetramer:RRM2B GeneArrowR-HSA-5632887 (Reactome)
TP53 Tetramer:RRM2B GeneArrowR-HSA-5632892 (Reactome)
TP53 Tetramer:SCO2 GeneArrowR-HSA-5632759 (Reactome)
TP53 Tetramer:SCO2 GeneArrowR-HSA-5632766 (Reactome)
TP53 Tetramer:TIGAR GeneArrowR-HSA-5628899 (Reactome)
TP53 Tetramer:TIGAR GeneArrowR-HSA-5628901 (Reactome)
TP53 TetramerR-HSA-5628899 (Reactome)
TP53 TetramerR-HSA-5629187 (Reactome)
TP53 TetramerR-HSA-5632759 (Reactome)
TP53 TetramerR-HSA-5632887 (Reactome)
TP53 TetramerR-HSA-5632914 (Reactome)
TP53 TetramerR-HSA-5632939 (Reactome)
TP63 Tetramer/ TP53 TetramerR-HSA-5632393 (Reactome)
TP63/T53:DDIT4 GeneArrowR-HSA-5632386 (Reactome)
TP63/T53:DDIT4 GeneArrowR-HSA-5632393 (Reactome)
TSC1:TSC2ArrowR-HSA-165179 (Reactome)
TSC1:TSC2R-HSA-380927 (Reactome)
TSC1:p-S1387-TSC2ArrowR-HSA-380927 (Reactome)
TSC1:p-S1387-TSC2ArrowR-HSA-380979 (Reactome)
TSC1R-HSA-165179 (Reactome)
TSC2R-HSA-165179 (Reactome)
TSC2R-HSA-198609 (Reactome)
TXNArrowR-HSA-73646 (Reactome)
TXNR-HSA-3341343 (Reactome)
mTORC1:Ragulator:Rag:GNP:RHEB:GDPArrowR-HSA-380979 (Reactome)
miR-26A RISCR-HSA-2318752 (Reactome)
p-AMPK heterotrimer:AMPR-HSA-5631941 (Reactome)
p-AMPK heterotrimer:AMPmim-catalysisR-HSA-380927 (Reactome)
p-S939,T1462-TSC2:14-3-3 dimerArrowR-HSA-5632732 (Reactome)
p-S939,T1462-TSC2ArrowR-HSA-198609 (Reactome)
p-S939,T1462-TSC2R-HSA-5632732 (Reactome)
p-T,p-S-AKTmim-catalysisR-HSA-198609 (Reactome)

Personal tools