Oncometabolites: tailoring our genes

Stefan Nowicki, Eyal Gottlieb, Stefan Nowicki, Eyal Gottlieb

Abstract

Increased glucose metabolism in cancer cells is a phenomenon that has been known for over 90 years, allowing maximal cell growth through faster ATP production and redistribution of carbons towards nucleotide, protein and fatty acid synthesis. Recently, metabolites that can promote tumorigeneis by altering the epigenome have been identified. These 'oncometabolites' include the tricarboxylic acid cycle metabolites succinate and fumarate, whose levels are elevated in rare tumours with succinate dehydrogenase and fumarate hydratase mutations, respectively. 2-Hydroxyglutarate is another oncometabolite; it is produced de novo as a result of the mutation of isocitrate dehydrogenase, and is commonly found in gliomas and acute myeloid leukaemia. Interestingly, the structural similarity of these oncometabolites to their precursor metabolite, α-ketoglutarate, explains the tumorigenic potential of these metabolites, by competitive inhibition of a superfamily of enzymes called the α-ketoglutarate-dependent dioxygenases. These enzymes utilize α-ketoglutarate as a cosubstrate, and are involved in fatty acid metabolism, oxygen sensing, collagen biosynthesis, and modulation of the epigenome. They include enzymes that are involved in regulating gene expression via DNA and histone tail demethylation. In this review, we will focus on the link between metabolism and epigenetics, and how we may target oncometabolite-induced tumorigenesis in the future.

Keywords: cancer; dioxygenase; epigenetics; metabolism; oncometabolites.

© 2015 The Authors. FEBS Journal published by John Wiley & Sons Ltd on behalf of FEBS.

Figures

Figure 1
Figure 1
Methyl and acetyl transfer pathways. The blue pathway depicts one‐carbon metabolism and its generation of SAM, which provides a methyl group for histone and DNA methylation. Imported folate is reduced to tetrahydrofolate (THF) and subsequently methylated to mTHF by the conversion of serine to glycine and the glycine cleavage system. The folate cycle is coupled to the methionine cycle by mTHF, donating a carbon to homocysteine, converting it to methionine. Adenylation of methionine produces SAM, which acts as a cosubstrate for DNMT and HMT, allowing transfer of its methyl group to DNA and histone tails respectively. The red pathway depicts acetyl transfer from acetyl‐coenzyme A. Acetyl‐coenzyme A, which is derived from pyruvate, links glycolysis to the TCA cycle, but is confined to the mitochondria. In the cytoplasm and nucleus, acetyl‐coenzyme A has to be derived by two alternative methods: first by ACLY, which utilizes citrate from the mitochondrial TCA cycle, and second by ACSS2, which ligates acetate to CoA. Acetyl‐coenzyme A can then be utilized as a cosubstrate by HAT, allowing transfer of the acetyl group to lysines on histone tails. 3‐PG, 3‐phosphoglyceric acid; SAH, S‐adenosylhomocysteine.
Figure 2
Figure 2
αKG‐dependent dioxygenases. This superfamily of enzymes uses oxygen and αKG as cosubstrates, resulting in the hydroxylation of the primary substrate and the decarboxylation of αKG, producing succinate and CO 2. These enzymes can be inhibited by elevated levels of 2HG, succinate, or fumarate. This occurs by competition with the cosubstrate, αKG, and/or by product inhibition, as for succinate.
Figure 3
Figure 3
Metabolic structure of oncometabolites: Succinate, fumarate and 2HG are closely linked, both structurally and metabolically, to αKG. Succinate and fumarate differ from αKG only by the presence of a hydroxyl group on C2 and the loss of C1. In addition, succinate and fumarate differ only by the presence of an ethylenic bond, which may explain their similar tumorigenic effects. 2HG differs from αKG and glutamate only by the presence of a hydroxyl group instead of a ketone group or amine group, respectively. This explains how 2HG can competitively inhibit αKG by occupying the same enzymatic binding site.

References

    1. Warburg O (1956) On the origin of cancer cells. Science 123, 309–314.
    1. Vander Heiden MG, Cantley LC & Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033.
    1. Ward PS & Thompson CB (2012) Metabolic reprogramming: a cancer hallmark even warburg did not anticipate. Cancer Cell 21, 297–308.
    1. Ben‐Haim S & Ell P (2009) 18F‐FDG PET and PET/CT in the evaluation of cancer treatment response. J Nucl Med 50, 88–99.
    1. Higashi K, Ueda Y, Yagishita M, Arisaka Y, Sakurai A, Oguchi M, Seki H, Nambu Y, Tonami H & Yamamoto I (2000) FDG PET measurement of the proliferative potential of non‐small cell lung cancer. J Nucl Med 41, 85–92.
    1. Jacob R, Welkoborsky HJ, Mann WJ, Jauch M & Amedee R (2001) [Fluorine‐18]fluorodeoxyglucose positron emission tomography, DNA ploidy and growth fraction in squamous‐cell carcinomas of the head and neck. ORL J Otorhinolaryngol Relat Spec 63, 307–313.
    1. Avril N, Menzel M, Dose J, Schelling M, Weber W, Janicke F, Nathrath W & Schwaiger M (2001) Glucose metabolism of breast cancer assessed by 18F‐FDG PET: histologic and immunohistochemical tissue analysis. J Nucl Med 42, 9–16.
    1. Buck AC, Schirrmeister HH, Guhlmann CA, Diederichs CG, Shen C, Buchmann I, Kotzerke J, Birk D, Mattfeldt T & Reske SN (2001) Ki‐67 immunostaining in pancreatic cancer and chronic active pancreatitis: does in vivo FDG uptake correlate with proliferative activity? J Nucl Med 42, 721–725.
    1. Kondoh H, Lleonart ME, Nakashima Y, Yokode M, Tanaka M, Bernard D, Gil J & Beach D (2007) A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid Redox Signal 9, 293–299.
    1. O'Neill LA & Hardie DG (2013) Metabolism of inflammation limited by AMPK and pseudo‐starvation. Nature 493, 346–355.
    1. Trabold O, Wagner S, Wicke C, Scheuenstuhl H, Hussain MZ, Rosen N, Seremetiev A, Becker HD & Hunt TK (2003) Lactate and oxygen constitute a fundamental regulatory mechanism in wound healing. Wound Repair Regen 11, 504–509.
    1. Anastasiou D, Poulogiannis G, Asara JM, Boxer MB, Jiang JK, Shen M, Bellinger G, Sasaki AT, Locasale JW, Auld DS et al (2011) Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 334, 1278–1283.
    1. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC et al (2009) Cancer‐associated IDH1 mutations produce 2‐hydroxyglutarate. Nature 462, 739–744.
    1. Jones PA & Baylin SB (2007) The epigenomics of cancer. Cell 128, 683–692.
    1. Feinberg AP & Vogelstein B (1983) Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 301, 89–92.
    1. Sandoval J & Esteller M (2012) Cancer epigenomics: beyond genomics. Curr Opin Genet Dev 22, 50–55.
    1. Noushmehr H, Weisenberger DJ, Diefes K, Phillips HS, Pujara K, Berman BP, Pan F, Pelloski CE, Sulman EP, Bhat KP et al (2010) Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17, 510–522.
    1. Toyota M, Ahuja N, Ohe‐Toyota M, Herman JG, Baylin SB & Issa JP (1999) CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A 96, 8681–8686.
    1. Rothbart SB & Strahl BD (2014) Interpreting the language of histone and DNA modifications. Biochim Biophys Acta 1839, 627–643.
    1. Verrier L, Vandromme M & Trouche D (2011) Histone demethylases in chromatin cross‐talks. Biol Cell 103, 381–401.
    1. Zhou VW, Goren A & Bernstein BE (2011) Charting histone modifications and the functional organization of mammalian genomes. Nat Rev Genet 12, 7–18.
    1. Seligson DB, Horvath S, Shi T, Yu H, Tze S, Grunstein M & Kurdistani SK (2005) Global histone modification patterns predict risk of prostate cancer recurrence. Nature 435, 1262–1266.
    1. Barlesi F, Giaccone G, Gallegos‐Ruiz MI, Loundou A, Span SW, Lefesvre P, Kruyt FA & Rodriguez JA (2007) Global histone modifications predict prognosis of resected non small‐cell lung cancer. J Clin Oncol 25, 4358–4364.
    1. Benard A, Goossens‐Beumer IJ, van Hoesel AQ, de Graaf W, Horati H, Putter H, Zeestraten EC, van de Velde CJ & Kuppen PJ (2014) Histone trimethylation at H3K4, H3K9 and H4K20 correlates with patient survival and tumor recurrence in early‐stage colon cancer. BMC Cancer 14, 531.
    1. Derissen EJ, Beijnen JH & Schellens JH (2013) Concise drug review: azacitidine and decitabine. Oncologist 18, 619–624.
    1. Slingerland M, Guchelaar HJ & Gelderblom H (2014) Histone deacetylase inhibitors: an overview of the clinical studies in solid tumors. Anticancer Drugs 25, 140–149.
    1. Dhanak D & Jackson P (2014) Development and classes of epigenetic drugs for cancer. Biochem Biophys Res Commun 455, 58–69.
    1. Locasale JW (2013) Serine, glycine and one‐carbon units: cancer metabolism in full circle. Nat Rev Cancer 13, 572–583.
    1. Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR & Thompson CB (2009) ATP‐citrate lyase links cellular metabolism to histone acetylation. Science 324, 1076–1080.
    1. Yoshii Y, Furukawa T, Yoshii H, Mori T, Kiyono Y, Waki A, Kobayashi M, Tsujikawa T, Kudo T, Okazawa H et al (2009) Cytosolic acetyl‐CoA synthetase affected tumor cell survival under hypoxia: the possible function in tumor acetyl‐CoA/acetate metabolism. Cancer Sci 100, 821–827.
    1. Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, Goodwin LM, Smethurst E, Mason S, Blyth K et al (2015) Acetyl‐CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 27, 57–71.
    1. Loenarz C & Schofield CJ (2008) Expanding chemical biology of 2‐oxoglutarate oxygenases. Nat Chem Biol 4, 152–156.
    1. 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.
    1. Gimm O, Armanios M, Dziema H, Neumann HP & Eng C (2000) Somatic and occult germ‐line mutations in SDHD, a mitochondrial complex II gene, in nonfamilial pheochromocytoma. Cancer Res 60, 6822–6825.
    1. Niemann S & Muller U (2000) Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet 26, 268–270.
    1. 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.
    1. Burnichon N, Briere JJ, Libe R, Vescovo L, Riviere J, Tissier F, Jouanno E, Jeunemaitre X, Benit P, Tzagoloff A et al (2010) SDHA is a tumor suppressor gene causing paraganglioma. Hum Mol Genet 19, 3011–3020.
    1. Alam NA, Barclay E, Rowan AJ, Tyrer JP, Calonje E, Manek S, Kelsell D, Leigh I, Olpin S & Tomlinson IP (2005) Clinical features of multiple cutaneous and uterine leiomyomatosis: an underdiagnosed tumor syndrome. Arch Dermatol 141, 199–206.
    1. Castro‐Vega LJ, Buffet A, De Cubas AA, Cascon A, Menara M, Khalifa E, Amar L, Azriel S, Bourdeau I, Chabre O et al (2014) Germline mutations in FH confer predisposition to malignant pheochromocytomas and paragangliomas. Hum Mol Genet 23, 2440–2446.
    1. Xiao M, Yang H, Xu W, Ma S, Lin H, Zhu H, Liu L, Liu Y, Yang C, Xu Y et al (2012) Inhibition of alpha‐KG‐dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev 26, 1326–1338.
    1. 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.
    1. Yan H, Parsons DW, Jin G, McLendon R, Rasheed BA, Yuan W, Kos I, Batinic‐Haberle I, Jones S, Riggins GJ et al (2009) IDH1 and IDH2 mutations in gliomas. N Engl J Med 360, 765–773.
    1. Parsons DW, Jones S, Zhang X, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Siu IM, Gallia GL et al (2008) An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812.
    1. Rakheja D, Konoplev S, Medeiros LJ & Chen W (2012) IDH mutations in acute myeloid leukemia. Hum Pathol 43, 1541–1551.
    1. Zou P, Xu H, Chen P, Yan Q, Zhao L, Zhao P & Gu A (2013) IDH1/IDH2 mutations define the prognosis and molecular profiles of patients with gliomas: a meta‐analysis. PLoS One 8, e68782.
    1. Borger DR, Goyal L, Yau T, Poon RT, Ancukiewicz M, Deshpande V, Christiani DC, Liebman HM, Yang H, Kim H et al (2014) Circulating oncometabolite 2‐hydroxyglutarate is a potential surrogate biomarker in patients with isocitrate dehydrogenase‐mutant intrahepatic cholangiocarcinoma. Clin Cancer Res 20, 1884–1890.
    1. Amary MF, Bacsi K, Maggiani F, Damato S, Halai D, Berisha F, Pollock R, O'Donnell P, Grigoriadis A, Diss T et al (2011) IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol 224, 334–343.
    1. Xu W, Yang H, Liu Y, Yang Y, Wang P, Kim SH, Ito S, Yang C, Xiao MT, Liu LX et al (2011) Oncometabolite 2‐hydroxyglutarate is a competitive inhibitor of alpha‐ketoglutarate‐dependent dioxygenases. Cancer Cell 19, 17–30.
    1. Reitman ZJ, Jin G, Karoly ED, Spasojevic I, Yang J, Kinzler KW, He Y, Bigner DD, Vogelstein B & Yan H (2011) Profiling the effects of isocitrate dehydrogenase 1 and 2 mutations on the cellular metabolome. Proc Natl Acad Sci U S A 108, 3270–3275.
    1. Ohka F, Ito M, Ranjit M, Senga T, Motomura A, Motomura K, Saito K, Kato K, Kato Y, Wakabayashi T et al (2014) Quantitative metabolome analysis profiles activation of glutaminolysis in glioma with IDH1 mutation. Tumour Biol 35, 5911–5920.
    1. Majmundar AJ, Wong WJ & Simon MC (2010) Hypoxia‐inducible factors and the response to hypoxic stress. Mol Cell 40, 294–309.
    1. 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.
    1. Zhao S, Lin Y, Xu W, Jiang W, Zha Z, Wang P, Yu W, Li Z, Gong L, Peng Y et al (2009) Glioma‐derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF‐1alpha. Science 324, 261–265.
    1. Williams SC, Karajannis MA, Chiriboga L, Golfinos JG, von Deimling A & Zagzag D (2011) R132H‐mutation of isocitrate dehydrogenase‐1 is not sufficient for HIF‐1alpha upregulation in adult glioma. Acta Neuropathol 121, 279–281.
    1. Koivunen P, Lee S, Duncan CG, Lopez G, Lu G, Ramkissoon S, Losman JA, Joensuu P, Bergmann U, Gross S et al (2012) Transformation by the (R)‐enantiomer of 2‐hydroxyglutarate linked to EGLN activation. Nature 483, 484–488.
    1. Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Campos C, Fabius AW, Lu C, Ward PS et al (2012) IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483.
    1. Duncan CG, Barwick BG, Jin G, Rago C, Kapoor‐Vazirani P, Powell DR, Chi JT, Bigner DD, Vertino PM & Yan H (2012) A heterozygous IDH1R132H/WT mutation induces genome‐wide alterations in DNA methylation. Genome Res 22, 2339–2355.
    1. Wang P, Dong Q, Zhang C, Kuan PF, Liu Y, Jeck WR, Andersen JB, Jiang W, Savich GL, Tan TX et al (2013) Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas. Oncogene 32, 3091–3100.
    1. Letouze E, Martinelli C, Loriot C, Burnichon N, Abermil N, Ottolenghi C, Janin M, Menara M, Nguyen AT, Benit P et al (2013) SDH mutations establish a hypermethylator phenotype in paraganglioma. Cancer Cell 23, 739–752.
    1. Figueroa ME, Abdel‐Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF et al (2010) Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–567.
    1. Losman JA, Looper RE, Koivunen P, Lee S, Schneider RK, McMahon C, Cowley GS, Root DE, Ebert BL & Kaelin WG Jr (2013) (R)‐2‐hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible. Science 339, 1621–1625.
    1. Chowdhury R, Yeoh KK, Tian YM, Hillringhaus L, Bagg EA, Rose NR, Leung IK, Li XS, Woon EC, Yang M et al (2011) The oncometabolite 2‐hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep 12, 463–469.
    1. Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel‐Wahab O, Edwards CR, Khanin R, Figueroa ME, Melnick A et al (2012) IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature 483, 474–478.
    1. Venneti S, Felicella MM, Coyne T, Phillips JJ, Gorovets D, Huse JT, Kofler J, Lu C, Tihan T, Sullivan LM et al (2013) Histone 3 lysine 9 trimethylation is differentially associated with isocitrate dehydrogenase mutations in oligodendrogliomas and high‐grade astrocytomas. J Neuropathol Exp Neurol 72, 298–306.
    1. Cedar H & Bergman Y (2009) Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 10, 295–304.
    1. Terunuma A, Putluri N, Mishra P, Mathe EA, Dorsey TH, Yi M, Wallace TA, Issaq HJ, Zhou M, Killian JK et al (2014) MYC‐driven accumulation of 2‐hydroxyglutarate is associated with breast cancer prognosis. J Clin Invest 124, 398–412.
    1. Fan J, Teng X, Liu L, Mattaini KR, Looper RE, Vander Heiden MG & Rabinowitz JD (2015) Human phosphogl‐ycerate dehydrogenase produces the oncometabolite d‐2‐hydroxyglutarate. ACS Chem Biol 10, 510–516.
    1. Nam H, Campodonico M, Bordbar A, Hyduke DR, Kim S, Zielinski DC & Palsson BO (2014) A systems approach to predict oncometabolites via context‐specific genome‐scale metabolic networks. PLoS Comput Biol 10, e1003837.
    1. Rohle D, Popovici‐Muller J, Palaskas N, Turcan S, Grommes C, Campos C, Tsoi J, Clark O, Oldrini B, Komisopoulou E et al (2013) An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 340, 626–630.
    1. Wang F, Travins J, DeLaBarre B, Penard‐Lacronique V, Schalm S, Hansen E, Straley K, Kernytsky A, Liu W, Gliser C et al (2013) Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 340, 622–626.
    1. Kernytsky A, Wang F, Hansen E, Schalm S, Straley K, Gliser C, Yang H, Travins J, Murray S, Dorsch M et al (2015) IDH2 mutation induced histone and DNA hypermethylation is progressively reversed by small molecule inhibition. Blood 125, 296–303.
    1. Bralten LB, Kloosterhof NK, Balvers R, Sacchetti A, Lapre L, Lamfers M, Leenstra S, de Jonge H, Kros JM, Jansen EE et al (2011) IDH1 R132H decreases proliferation of glioma cell lines in vitro and in vivo. Ann Neurol 69, 455–463.
    1. Tonjes M, Barbus S, Park YJ, Wang W, Schlotter M, Lindroth AM, Pleier SV, Bai AH, Karra D, Piro RM et al (2013) BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild‐type IDH1. Nat Med 19, 901–908.

Source: PubMed

3
Abonnere