Nrc 3884Artigo

download Nrc 3884Artigo

of 15

Transcript of Nrc 3884Artigo

  • 8/9/2019 Nrc 3884Artigo

    1/15

    -N-methyl-lysine was first found in a bacterial flagel-lar protein in 1959 (REF. 1)and, 5 years later, this post-translational modification (PTM) was also identified inhistone proteins2. Biochemically, S-adenosyl-L-methionine(AdoMet) is the principal methyl donor in the methyl-transferase reaction and is the second most widely usedenzyme substrate following ATP3. This frequency indi-cates the importance of the methylation reaction in

    various biochemical or metabolic pathways. Althoughthe physiological importance of protein lysine methyla-tion was unknown for many years, several protein lysinemethyltransferases (PKMTs) have now been identified,and their physiological significance, particularly inthe field of epigenetics, has begun to be elucidated48.The SET-domain proteins constitute a major group of

    AdoMet-dependent PKMTs; nearly 50 human proteinsare categorized into this family, although not all of themhave known PKMT activity. In addition to the SET-domain proteins, several non-SET-domain proteins,including DOT1-like histone H3K79 methyltransferase(DOT1L), are also reported to have PKMT activity9.

    Moreover, protein lysine methylation was thought tobe irreversible because the half-life of the histone meth-ylation was approximately equivalent to the half-life ofhistones themselves10. However, the first protein lysinedemethylase lysine-specific demethylase 1 (LSD1; alsoknown as KDM1A) was discovered in 2004 (REF. 11), andsubsequently the Jumonji C (JmjC)-domain-containing

    family was reported to have protein demethylase activ-ity12. These findings indicate that protein lysine methyla-tion seems to be dynamically regulated.

    Many reports have indicated that dysregulation ofPKMTs and protein lysine demethylases (PKDMs)has substantial roles in tumorigenesis4,13,14, and theseenzymes are considered to be important targets forthe development of anticancer therapy15,16. Functionalanalyses of PKMTs and PKDMs have been carried outprimarily in the context of their role in epigenetic regu-lation. For instance, the PKMT EZH2, which methyl-ates histone H3 at lysine 27 (H3K27), is a componentof Polycomb-repressive complexes and is overexpressedin various types of cancer17. Polycomb-repressive com-plexes play a fundamental part in controlling the expres-

    sion of downstream genes, including cyclin-dependentkinase inhibitor 2A (CDKN2A), to promote cell cycleprogression in physiological and pathological condi-tions18. Similarly, several reports have demonstratedthat aberrations of other PKMTs and PKDMs promotemalignant transformation via histone methylation-dependent transcriptional regulation4,13,1921.

    Beyond histones, the biological and physiologicalsignificance of non-histone lysine methylation in humantumorigenesis has recently begun to be explored2227.Accumulating evidence indicates that, similar to otherPTMs such as phosphorylation and acetylation, lysinemethylation may be important not only in epigenetic

    Section of Hematology/

    Oncology, Department of

    Medicine, The University of

    Chicago, 5841 S. Maryland

    Avenue, MC 2115 Chicago,

    Illinois 60637, USA.

    Correspondence to R.H.

    e-mail: rhamamoto@

    medicine.bsd.uchicago.edu

    doi:10.1038/nrc3884

    S-adenosyl-L-methionine

    (AdoMet).A moleculesynthesized from methionine

    and ATP by methionine adeno-

    syltransferase. The methylation

    group attached to the

    methionine sulphur atom in

    AdoMet is chemically reactive.

    This allows donation of this

    group to an acceptor substrate

    in transmethylation reactions.

    Critical roles of non-histoneprotein lysine methylation inhuman tumorigenesisRyuji Hamamoto, Vassiliki Saloura and Yusuke Nakamura

    Abstract | Several protein lysine methyltransferases and demethylases have been identified

    to have critical roles in histone modification. A large body of evidence has indicated that

    their dysregulation is involved in the development and progression of various diseases,including cancer, and these enzymes are now considered to be potential therapeutic targets.

    Although most studies have focused on histone methylation, many reports have revealed

    that these enzymes also regulate the methylation dynamics of non-histone proteins such as

    p53, RB1 and STAT3 (signal transducer and activator of transcription 3), which have

    important roles in human tumorigenesis. In this Review, we summarize the molecular

    functions of protein lysine methylation and its involvement in human cancer, with a particular

    focus on lysine methylation of non-histone proteins.

    REVIEWS

    110 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

    mailto:[email protected]:[email protected]:[email protected]:[email protected]
  • 8/9/2019 Nrc 3884Artigo

    2/15

  • 8/9/2019 Nrc 3884Artigo

    3/15

    Competitiveinhibition

    Indirect effect

    on othermodifications

    Cytoplasm

    Methylated-lysine-specificbinding proteins

    Inhibition of

    proteinproteininteractions

    Inhibition ofpolyubiquitylation

    Protein degradationby proteasomes

    Transcriptionfactors

    Methylationactivatestranscription

    a Other protein modifications b Proteinprotein interactions c Protein stability

    d e Promoter binding

    K Me KMeMe

    KMeMeUb

    Ub UbUb Ub Ub

    Ac

    K SMe P

    Chromodomain

    KMeMe

    UbUbUb

    Ub Ub

    HSP70

    HSP70

    HSP70

    Subcellularlocalization

    Me Me Me

    AURKB

    Activate

    Nucleus

    Promoter

    Oxygenase

    An enzyme of the

    oxidoreductase class that

    catalyses the incorporation of

    both atoms of molecular

    oxygen into the substrate.

    energies and hydrogen bonding potential of the lysineside chains. Indeed, there are methyl lysine-binding pro-teins that have a special motif such as a chromodomainfor recognizing methylated lysine residues, called thearomatic cage. This aromatic cage comprises a collec-tion of aromatic protein residues, often accompanied byone or more neighbouring anionic residues34. The com-bination of favourable cation-, electrostatic and vander Waals interactions, as well as shape complementa-rity, provides methyl lysine-binding proteins with a highdegree of specificity for methylated lysine35. By contrast,lysine methylation of MAPK kinase kinase 2 (MAP3K2)inhibits its interaction with the serine/threonine proteinphosphatase 2A (PP2A) complex, which is a key negativeregulator of the MAPK pathway, implying that methyllysine can also block proteinprotein interactions33.

    Protein stability.As lysine polyubiquitylation plays akey part in protein degradation through the ubiquitinproteasome pathway, lysine methylation may increasethe stability of proteins by preventing polyubiquityla-tion.Indeed, in Saccharomyces cerevisiae, lysine methyl-ated proteins show a significantly longer half-life thannon-methylated proteins. Moreover, 43% of methylatedlysine sites are predicted to be amenable to ubiquityla-tion, suggesting that methylated lysine residues mightblock the action of ubiquitin ligases36(FIG. 2c). However,the DCAF1DDB1CUL4 E3 ubiquitin ligase complexrecognizes monomethylated lysine and promotes poly-ubiquitylation of the other lysine residues on substrates

    such as retinoic acid-related orphan nuclear receptor-(ROR)37, implying that lysine methylation may alsodestabilize target proteins through regulation of distantpolyubiquitylation.

    Subcellular localization.Typically, a nuclear localiza-tion signal consists of one or more short sequences ofpositively charged lysine or arginine residues exposedon the protein surface38. As both lysine and arginineresidues are critical for the nuclear localization of pro-teins, one could speculate that methylation of lysine orarginine may affect subcellular localization. Indeed,some lysine methylated proteins such as heat shock pro-tein 70 (HSP70) and p53 are predominantly localizedin the nucleus, whereas unmodified versions of thesesame proteins are localized in both the cytoplasm andthe nucleus23,39(FIG. 2d)

    Promoter binding.Lysine methylation also regulatesthe binding affinity of transcription factors for pro-moters, which changes the transcription levels of tar-get genes40(FIG. 2e). For example, lysine methylation ofp53 by the PKMT SETD7 and that of nuclear factor-B(NF-B) by the PKMT nuclear receptor-bindingSET domain-containing protein 1 (NSD1) markedlyincreased their promoter binding ability and activationof downstream genes39,41. In a structural modellinganalysis of RELA (a subunit of NF-B) in complex withDNA, two methylated lysine residues on RELA wereshown to interact with DNA through hydrophobic

    Figure 2 | Molecular functions of lysine methylation in human tumorigenesis. The biological importance of lysine

    methylation is primarily categorized into five groups. a | Lysine methylation affects other protein modifications directly

    (through competitive inhibition) or indirectly.b | Lysine methylation regulates proteinprotein interactions. Methylated-

    lysine-specific binding proteins have been reported, and these proteins contain motifs that specifically recognizemethylated lysine residues such as chromodomains and Tudor domains. Methylated lysine can also negatively regulate

    some proteinprotein interactions. c | Lysine methylation competitively inhibits the polyubiquitylation of lysine residues

    and stabilizes the protein. Additionally, lysine methylation also promotes the polyubiquitylation of other lysine residues on

    the same protein, leading to protein destabilization.d | Subcellular localization is regulated by lysine methylation. For

    example, methylated heat shock protein 70 (HSP70) proteins are predominantly localized in the nucleus, whereas

    unmodified versions are predominantly localized in the cytoplasm. e | Lysine methylation regulates promoter binding

    affinity of transcription factors, thereby changing transcription levels of target genes. Ac, acetyl group; AURKB, Aurora

    kinase B; P, phosphate group; Me, methyl group; Ub, ubiquitin.

    REVIEWS

    112 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    4/15

    Tudor domains

    Protein domains originally

    identified as a region of 50

    amino acids found in the

    Drosophila melanogaster

    Tudor protein. The structurally

    characterized Tudor domain in

    human proteins recognizes

    symmetrically dimethylated

    arginine. This domain is also

    reported as a methyl

    lysine-binding protein module.

    contacts42. This result reveals that increased hydropho-bicity of lysine residues by methylation can enhancethe promoter binding ability of transcription factors.

    Non-histone PKMT and PKDM substrates

    So far, nearly 20 non-histone substrates have been dis-covered for PKMTs and PKDMs (TABLE 1). Many PKMTsand PKDMs are localized in the nucleus, and severalnuclear proteins, including transcription factors, canserve as substrates of PKMTs and PKDMs. However,several PKMTs or PKDMs are also localized in the cyto-plasm, and cytoplasmic proteins are also reported tobe substrates of these enzymes. Below, we highlight thelysine methylation of non-histone proteins involved inseveral tumour-associated signalling pathways.

    p53. p53 is one of the most important tumour sup-pressor genes involved in human tumorigenesis43.Chuikov et al.39demonstrated that SETD7 methyl-ates lysine 372 of p53 (p53K372) and that this meth-ylation enhances p53 stability and transcriptional

    activity. K372-methylated p53 is restricted to thenucleus, although p53 is equally distributed betweenthe nuclear and cytosolic f ractions39. Subsequently,Huang et al.44 showed that the PKMT SET andMYND-domain containing 2 (SMYD2) monometh-ylates lysine 370 of p53 (p53K370); this lysine residueis located in the regulatory domain at the extremecarboxyl terminus of p53 (FIG. 3a). Knockdownof SMYD2 by small interfering RNA enhancesp53-mediated apoptosis in cancer cells44. In addition,SMYD2-dependent p53K370 methylation impairsthe expression of CDKN1A, an important down-stream target of p53, implying that SMYD2 repressesthe function of p53 through K370 monomethylation.Moreover, p53K372 methylation by SETD7 seemsto inhibit SMYD2-depedent p53K370 methylationthrough blocking the interaction between SMYD2and p53 (REFS 39,44)(FIG. 3a).

    The function of monomethylation and dimethylationof p53 on lysine 370 seems to be different. For example,LSD1 was reported to demethylate p53K370 and repressp53 activity45(FIG. 3a). In this case, LSD1 preferentiallyreverses dimethylated p53K370 in cancer cells, althoughit can remove both monomethylation and dimethylationat lysine 370 in biochemical reactions in vitro45. p53K370dimethylation promotes the association of p53 with theco-activator p53-binding protein 1 (p53BP1) through

    tandem Tudor domainsin p53BP1 (REF. 45), leading toincreased p53 function, including apoptosis induc-tion. These results indicate that monomethylation ofp53K370 is crucial for the repression of p53 activity,whereas dimethylation of p53K370 seems to activatep53. The complexity of methylation pathways in humancancer is highlighted by the fact that both SMYD2 andLSD1 affect the same lysine residue of p53, and thatboth of these enzymes can promote oncogenesis and areoverexpressed in various types of cancer22,46. It is plau-sible that their oncogenic actions on p53 may be syn-ergistic, although they could also have effects on othercancer-related pathways.

    Furthermore, Shi et al.47reported that the proteinlysine methyltransferase SETD8 monomethylates p53at lysine 382 and that this methylation suppressesp53-dependent transcription activation in cancercells. Taken together, these results indicate that lysinemethylation is an important regulator of p53 function.

    RB1 and E2F.RB1 is a key cell cycle regulator and tumoursuppressor, and is dysfunctional in several cancer types48.RB1 was identified as a binding partner for mitogeniconcoproteins49and was also discovered to be phosphoryl-ated in synchrony with the cell cycle50. Recently, we andothers reported that lysine methylation of RB1 seems tobe one of the substantial regulators of RB1 function and ispivotal for cell cycle regulation22,51,52. Methylation at lysine810 of RB1 by SMYD2 is likely to enhance phosphory-lation of RB1 at serines 807 and 811 (REF. 22). Additionally,although RB1 normally interacts with E2F (a family oftranscription factors that modulate important cellularevents, including cell cycle progression, apoptosis andDNA damage response53) to suppress transcription of

    E2F target genes, lysine 810 methylation of RB1 acceler-ates E2F transcriptional activity through enhancement ofRB1 phosphorylation and promotes cell cycle progression(FIG. 3b). Furthermore, we identified that lysine 442 of pro-tein phosphatase 1, regulatory subunit 12A (PPP1R12A;also called MYPT1) in the myosin phosphatase holo-enzyme, which stimulates dephosphorylation of RB1, isa target for methylation and demethylation catalysed bySETD7 and LSD1, respectively24

    (FIG. 3b). Demethylationof PPP1R12A by LSD1 enhances PPP1R12A polyubiq-uitylation, which increases the proteasome-mediateddegradation of PPP1R12A and therefore the amount ofphosphorylated RB1. Subsequently, released E2F acti-

    vates transcription genes required for S phase, and cellcycle progression is enhanced24. Together, methylationand demethylation dynamics seems to play an impor-tant part in cell cycle progression through the regulationof RB1 activity.

    Deregulated expression or activity of membersof the E2F family has also been detected in manyhuman cancers54. Kontaki et al.55demonstrated that inp53-deficient cancer cells, methylation of lysine 185 ofE2F1 (E2F1K185) by SETD7 inhibits acetylation andphosphorylation at distant positions and, in parallel,induces polyubiquitylation and degradation of E2F1.This process prevents E2F1 accumulation duringDNA damage and activation of its pro-apoptotic tar-

    get gene TP73(FIG. 3b). Moreover, LSD1 demethylatesE2F1K185, which is important to maintain a substan-tial pool of unmethylated E2F1 in cancer cells, whichcan then be subjected to lysine acetyltransferase 2B(KAT2B)-mediated hyperacetylation, as well as check-point kinase 2 (CHEK2)-mediated phosphorylation,following DNA damage55(FIG. 3b). These results implythat in p53-deficient cancer cells, LSD1 and SETD7can influence DNA damage-induced cell death in amanner that is in contrast to the aforementionedp53-dependent apoptosis induction regulated byLSD1 and SETD7. These results suggest that antican-cer treatments that combine DNA-damaging agents

    REVIEWS

    NATURE REVIEWS |CANCER VOLUME 15 |FEBRUARY 2015 |113

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    5/15

    Table 1 | Protein lysine methyltransferases and demethylases involved in human tumorigenesis and their substrates

    Family Enzymename (alias)

    Domain Subcellularlocalization

    Substrate Cancer type Refs

    Histone Non-histone

    Lysine methyltransferases

    Polycombcomplex

    EZH2 (KMT6) SET, CXC andSANT

    Nucleus*and

    cytoplasm

    H3K27 andH2BK120

    RORand STAT3 AML, bladder cancer, breastcancer, CCC, CML, CRC,

    glioblastoma, lymphoma,NSCLC, oesophageal cancer,osteosarcoma, SCLC and RCC

    17,31,37,74

    SMYDfamily

    SMYD2(KMT3C)

    SET Cytoplasm*and nucleus

    H3K4 andH3K36

    p53, RB1, PARP1,HSP90AB1 and ER

    Bladder cancer, breastcancer, cervical cancer, CRC,HCC, head and neck cancer,lymphoma, oesophagealcancer, ovarian cancer,pancreatic cancer, prostatecancer, seminoma and skincancer

    22,25,27,44,51,52,69,111113

    SMYD3(KMT3E)

    SET Cytoplasm*and nucleus

    H3K4 andH4K5

    VEGFR1 andMAP3K2

    ACC, breast cancer, CCC,cervical cancer, CRC,HCC, lung cancer, MTC,oesophageal cancer,pancreatic and prostatecancer

    4,13,26,33,114121

    NSD family NSD1(KMT3B)

    SET, PWWP,AWS, PHD,RING andPostSET

    Nucleus*andchromosome

    H3K36 NF-B AML, glioblastoma, lungcancer and multiple myeloma

    122124

    WHSC1(MMSET andNSD2)

    SET, PWWP,AWS, PHD,RING andPostSET

    Nucleus*,cytoplasmandchromosome

    H3K36 Bladder cancer, breastcancer, CCC, CML, HCC,multiple myeloma, NSCLC,oesophageal cancerosteosarcoma, prostatecancer, RCC and SCLC

    125129

    WHSC1L1(NSD3)

    SET, PWWP,AWS, PHD,RING and

    PostSET

    Nucleus*andchromosome

    H3K36 Bladder cancer, breast cancer,lymphoma and SCLC

    130132

    SETDfamily

    SETD1A(KMT2F)

    SET, N-SET andPRM

    Nucleus*andchromosome

    H3K4 HSP70 Bladder cancer, CRC, HCC,lung cancer and RCC

    23,133

    SETD7(KMT7)

    SET and MORN Nucleus*andchromosome

    H3K4 PPP1R12A, p53,NF-B, E2F1, DNMT1and STAT3

    Breast cancer and multiplemyeloma

    24,39,55,66,73,134,135

    SETD8(KMT5A)

    SET Nucleus*andchromosome

    H4K20 PCNA and p53 Bladder cancer, CML, HCC,NSCLC and SCLC

    47,75

    SUV39family

    SUV39H2(KMT1B)

    SET, PreSET,PostSET andchromodomain

    Nucleus*andchromosome

    H3K9 andH2AXK134

    Bladder cancer, cervicalcancer, NSCLC, oesophagealcancer, osteosarcoma,prostate cancer and STT

    32,136

    EHMTfamily

    EHMT2(KMT1C)

    SET, PreSET,PostSET andANK

    Nucleus*andchromosome

    H3K9 C/EBP AML, bladder cancer, breastcancer, CCC, CML, NSCLC,oesophageal cancer, prostatecancer and SCLC

    19,71,137,138

    MLL family MLL2(KMT2D)

    SET, PHD, RING,FYRN, FYRC andHMG

    Nucleus* H3K4 Bladder cancer, breast cancer,CRC, lung cancer, melanomaand MLL

    139

    MLL3(KMT2C)

    SET, PHD, RING,FYRN, FYRC,HMG, AT_hook,C1 and PostSET

    Nucleus* H3K4 Breast cancer, glioblastoma,melanoma, MLL, oesophagealcancer, pancreatic cancer andstomach cancer

    140142

    DOT1Lfamily

    DOT1L(KMT4)

    AT_hook Nucleus* H3K79 MLL 143145

    REVIEWS

    114 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    6/15

    Family Enzymename (alias)

    Domain Subcellularlocalization

    Substrate Cancer type Refs

    Histone Non-histone

    Lysine demethylases

    LSD1family

    LSD1(KDM1A)

    FADbinding 2and SWIRM

    Nucleus* H3K4 andH3K9

    PPP1R12A, p53, ERand STAT3

    Bladder cancer, CRC andSCLC

    11,24,45,46

    JMJDfamily

    JMJD1A(KDM3A)

    JmjC Nucleus*andcytoplasm

    H3K9 Bladder cancer, CCC, HCC,NSCLC, osteosarcoma, SCLCand RCC

    20,146,147

    JMJD2A(KDM4A)

    JmjC, JmjN, PHDand Tudor

    Nucleus* H3K9 andH3K36

    Bladder cancer, breast cancer,CML, NSCLC, oesophagealcancer, osteosarcoma, ovariancancer, prostate cancer, SCLC,stomach cancer and uterinecancer

    148151

    JMJD2B(KDM4B)

    JmjC, JmjN, PHDand Tudor

    Nucleus* H3K9 Bladder cancer, CRC, NSCLC,oesophageal cancer, SCLC,stomach cancer and RCC

    21,152154

    JMJD3(KDM6B)

    JmjC Nucleus* H3K27 Glioblastoma 155

    UTX UTX(KDM6A)

    JmjC and TPR Nucleus* H3K27 Bladder cancer, breast cancer,CML, CRC, glioblastoma,multiple myeloma, NSCLC,oesophageal cancer,pancreatic cancer, RCC andSCLC and TALL

    156159

    JARIDfamily

    JARID1B(KDM5B)

    JmjC, JmjN,ARID, PHD,ZFC5HC2 andPLU-1

    Nucleus* H3K4 AML, bladder cancer, breastcancer, cervix cancer, CML,CRC, melanoma, NSCLC,oesophageal cancer, SCLCand RCC

    160164

    ACC, adenocarcinoma; AML, acute myeloid leukaemia; CCC, cholangiocarcinoma; C/EBP, CCAAT/enhancer binding protein; CML, chronic myelogenousleukaemia; CRC, colorectal cancer; DNMT1, DNA (cytosine5)methyltransferase 1; DOT1L, DOT1like histone H3K79 methyltransferase; EHMT, euchromatichistone-lysine N-methyltransferase; ER, oestrogen receptor-; H3K27, histone H3 lysine 27; HCC, hepatocellular carcinoma; HSP, heat shock protein; JARID,

    Jumonji, ATrich interactive domain; JMJD, Jumonji domaincontaining; KMT, lysineN-methyltransferase; LSD1, lysine-specific demethylase 1; MAP3K2, MAPKkinase kinase 2; MLL, mixedlineage leukaemia; MTC, medullary thyroid cancer; NFB, nuclear factor-B; NSCLC, nonsmall cell lung carcinoma; NSD, nuclearreceptorbinding SET domaincontaining; PARP1, poly(ADPribose) polymerase 1; PCNA, proliferating cell nuclear antigen; PPP1R12A, protein phosphatase 1,regulatory subunit 12A; RCC, renal cell carcinoma; ROR,retinoic acid-related orphan nuclear receptor-; SCLC, small cell lung carcinoma; SETD, SETdomain-containing; SMYD, SET and MYND-domain-containing; STAT3, signal transducer and activator of transcription 3; STT, soft tissue tumour; SUV39H,suppressor of variegation 39 homologue; T-ALL, T-cell acute lymphoblastic leukaemia; VEGFR1, vascular endothelial growth factor receptor 1.*The predominantsubcellular localization.

    Table 1 (cont.) | Protein lysine methyltransferases and demethylases that are involved in human tumorigenesis and their substrates

    and drugs modulating LSD1 or SETD7 activity mayhave inverse effects depending on the p53 status incancer cells.

    Heat shock proteins.HSPs are overexpressed in a widerange of human cancers and have substantial roles intumour cell proliferation, differentiation, invasion,metastasis and recognition by the immune system56.

    Increased expression of HSPs can also protect malignantcells from activation of pro-apoptotic signalling, and thismay underlie the role of HSPs in tumour progressionand resistance to treatment57. HSP70 and HSP90AB1undergo several PTMs, and we previously identified thatthey are also lysine methylated23,25.

    HSP70 is a ubiquitous molecular chaperone thatfunctions in a myriad of biological processes, includingmodulation of polypeptide folding, protein degrada-tion, subcellular translocation of proteins across mem-branes and proteinprotein interactions58. We reportedthat dimethylation of HSP70 lysine 561 was markedlyincreased in cancer cells, and methylated HSP70 was

    predominantly localized in the nucleus. This is in contrastto non-methylated HSP70, which is localized predomi-nantly in the cytoplasm23. The methylation of HSP70 iscatalysed by the PKMT SETD1A, which is overexpressedin various types of cancer23. The lysine 561 methylatedHSP70 preferentially binds to and activates Aurorakinase B in the nucleus, resulting in cell cycle progressionin cancer cells23(FIG. 2d). The importance of this modifica-

    tion in cancer was confirmed by immunohistochemicalanalysis using an HSP70K571 dimethylation-specificantibody, which revealed positive nuclear staining in 354of 409 (86.6%) non-small cell lung carcinoma cases. Inaddition to lung cancer, this positive staining of lysine561 methylated HSP70 was commonly found in bladderand kidney cancer tissues, whereas no such staining wasobserved in non-neoplastic tissues.

    HSP90 is an evolutionarily conserved molecularchaperone that participates in the stabilization and acti-

    vation of more than 200 proteins, which are referred to asHSP90 clients. Many of the client proteins of HSP90 areessential for various cell signalling pathways, including

    REVIEWS

    NATURE REVIEWS |CANCER VOLUME 15 |FEBRUARY 2015 |115

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    7/15

    a

    b

    c

    Me Me

    TA1 and TA2 PRR DBDL OD CTDp53

    370 KSKKGQSTSRHKK 382

    SMYD2

    SMYD2

    K370me1K370me2 toK370me1

    SETD7

    SETD7

    SETD8

    K372me1

    p53 inactivation Inhibition of K370me1

    K382me1

    p53 inactivation

    RB1

    CDK4

    High affinity

    E2F1

    K185

    SMYD2-dependent RB1methylation activates E2F

    Cell cycle progression

    Regulation of E2F1-dependentapoptosis in p53-deficient cancer cells

    p53

    53BP1

    Me

    Me

    Me

    Me

    Me

    Me

    Me

    MeMe

    p53

    53BP1LSD1

    LSD1

    LSD1

    SETD7 LSD1

    VEGFR1

    MAP3K2

    TM

    SMYD3

    K260

    PP2A

    Inhibition ofPP2A interaction Activation of oncogenic

    RAS signalling

    IG-LD

    PPP1R12A

    PPP1R12AdephosphorylatesRB1

    K442

    CC

    CTD

    K810

    Enhances RB1hyperphosphorylation

    K831me2

    K260me2or K260me3

    ANK

    Stabilizes PPP1R12A protein

    Pocket A Pocket B

    DBD CC MB TD

    TK1 TK2

    K831Activation ofkinase activity

    PB1 Kinase domain

    Figure 3 | Effects of lysine methylation on the pathways of p53, RB1 and protein kinases. a | Methylation of lysine

    at residues 370, 372 and 382 in the carboxyterminal domain (CTD) of p53 has been identified. Lysine 370dimethylated p53

    (p53K370me2) can promote the association of p53 with the coactivator p53binding protein 1 (53BP1) through tandem

    Tudor domains in 53BP1. By contrast, lysine-specific demethylase 1 (LSD1) prevents the accumulation of p53K370me2 by

    demethylating this site, thereby preventing the binding of 53BP1 to p53. Both LSD1 and the protein lysine

    methyltransferase (PKMT) SET and MYND-domain containing 2 (SMYD2) produce p53K370me1 that results in the

    inactivation of p53. Meanwhile, the PKMT SETD7 monomethylates K372 (K372me1), which inhibits K370me1 and activatesp53. SETD8 also inactivates p53 through monomethylation of K382 (K382me1). b | The RB1 pathway is regulated by lysine

    methylation. Lysine methylation sites on RB1, E2F1 and protein phosphatase 1, regulatory subunit 12A (PPP1R12A) are

    displayed. SETD7 monomethylates K442, which stabilizes PPP1R12A, and this methylation is reversibly demethylated by

    LSD1. SMYD2 monomethylates K810 on RB1 and increases the affinity between cyclindependent kinase 4 (CDK4) and RB1,

    which enhances hyperphosphorylation of RB1 and therefore activates E2F. SETD7 monomethylates K185 of E2F1

    (E2F1K185me1), which prevents apoptotic functions of E2F1 in p53-deficient cells during DNA damage, and E2F1K185me1

    is demethylated by LSD1, promoting E2F1 stability and apoptosis.c | K831 of vascular endothelial growth factor receptor 1

    (VEGFR1) and K260 of MAPK kinase kinase 2 (MAP3K2) are methylated by SMYD3. VEGFR1K831 dimethylation by SMYD3

    activates the kinase activity of VEGFR1. K260 of MAP3K2 is dimethylated and trimethylated by SMYD3, which activates

    oncogenic RAS signalling through inhibition of the MAP3K2PP2A (protein phosphatase 2A) interaction. ANK, ankyrin

    repeat; CC, coiledcoil domain; DBD, DNAbinding domain; IGLD, immunoglobulinlike domain; MB, markedbox;

    OD, oligomerization domain; PB1, Phox and Bem1p; PRR, prolinerich region; TA, transcriptionactivation domain;

    TD, transactivation domain; TK, tyrosine kinase domain; TM, transmembrane domain.

    REVIEWS

    116 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    8/15

    Nitrosylation

    Nitrosylation, specifically

    S-nitrosylation, involves the

    covalent incorporation of a

    nitric oxide moiety into thiol

    groups, to form S-nitrosothiol.

    S-nitrosylation is a

    physiologically important

    post-translational modification

    that affects a variety of

    proteins involved in a number

    of cellular processes.

    adaptive responses to various stresses57,59. HSP90 andproteins called co-chaperones form the dynamic com-plex known as the HSP90 chaperone machinery60.Cancer cells use the HSP90 chaperone machinery to pro-tect an array of mutated and overexpressed oncoproteinsfrom misfolding and degradation. Therefore, HSP90 isrecognized as a critical facilitator of oncogene addictionand cancer cell survival61. HSP90 function is regulated by

    various PTMs such as acetylation, phosphorylation andnitrosylation. We also reported methylation of lysines 531and 574 of HSP90AB1 by SMYD2. The lysine methyla-tion of HSP90AB1 is important for its homodimerizationand its interaction with stress-induced phosphoprotein 1(STIP1) and cell division cycle 37 (CDC37), which areco-chaperones of HSP90AB1 in human cancer cells,resulting in enhancement of cancer cell growth25.

    Protein kinases (VEGFR1 and MAP3K2). Vascularendothelial growth factor receptor 1 (VEGFR1), a recep-tor tyrosine kinase, mediates signalling that is involvedin cell proliferation and angiogenesis62. We found that

    SMYD3 methylates lysine 831 of VEGFR1, which islocated in the tyrosine kinase domain, and this meth-ylation enhances the kinase activity of VEGFR1 bothin vitroand in vivo26(FIG. 3c). VEGFR1 in cancer cellsis implicated in liver metastasis of colorectal cancer63,and exogenous expression of VEGFR1 enhances themigration and invasion of pancreatic cancer cells64.Hence, therapeutic approaches targeting VEGFR1 meth-ylation by SMYD3 can directly attenuate kinase activ-ity of VEGFR1 in cancer cells and benefit patients byinhibiting invasion and metastasis of cancer cells.

    SMYD3 can methylate MAP3K2, a member of the ser-ine/threonine protein kinase family33. SMYD3-mediatedmethylation of MAP3K2 at lysine 260 activates the RASRAFMEKERK signalling module, and SMYD3 deple-tion synergizes with a MEK inhibitor to block RAS-driventumorigenesis33(FIG. 3c). A clinical implication of this find-ing is the identification of SMYD3 as a candidate thera-peutic target to treat pancreatic and lung cancers drivenby RAS, as well as potentially other RAS-driven tumours.The complete loss of SMYD3 function has no apparentphenotype in mice, implying that SMYD3 inhibitorswould have minimal adverse effects as chemotherapeuticagents33. Consequently, we could envisage a therapeuticstrategy comprising inhibitors of RAF or MEK (whichare already currently used for anticancer therapy) withan SMYD3 inhibitor, which could mitigate potential side

    effects by lowering the overall dosage needed for eachagent and also limit the development of resistance.

    Transcription factors (NF-B, ER, C/EBP andSTAT3).The NF-B transcription factor regulates multi-ple biological functions, including inflammation, immu-nity, cell proliferation and apoptosis65. SETD7 methylateslysine 37 of RELA in the nucleus, which enhances thepromoter binding affinity of RELA. This methylationseems to have a critical role in the induction of a sub-set of NF-B-dependent genes in response to tumournecrosis factor- (TNF) stimulation66. In addition, Luet al.41showed that NSD1 methylates lysines 218 and 221

    of RELA and activates NF-B activity. Given that theK218A, K221A or combined K218A and K221A muta-tion of RELA substantially diminished its DNA-bindingability, it seems that methylation of several lysine residueson NF-B plays an important part in NF-B-dependenttranscriptional regulation through increasing its bind-ing to promoters of downstream genes. Interestingly,these two methylation sites of RELA are demethylatedby the PKDM F-box and leucine-rich repeat protein 11(FBXL11), and overexpression of FBXL11 inhibitsNF-B activation and retards the growth of HT29 coloncancer cells41(FIG. 4a). Furthermore, SETD6 methylatesRELA on lysine 310 (RELAK310), and this methyla-tion renders RELA inert and attenuates RELA-driventranscriptional programmes, including inflammatoryresponses in primary immune cells67. MonomethylatedRELAK310 is recognized by the ankyrin repeat ofeuchromatic histone-lysine N-methyltransferase 1(EHMT1), and this interaction stabilizes EHMT1 activ-ity. As EHMT1 works as a transcriptional repressorthrough generating monomethylated and dimethylated

    H3K9 at euchromatin, the interaction of EHMT1 withRELA attenuates transcription of NF-B target genes(FIG. 4a). Taken together, NF-B activity is intricatelyregulated by lysine methylation of RELA in a positiveor negative manner depending on the methylation site.

    Oestrogen receptor- (ER), a ligand-activatedtranscription factor involved in human breast cancer 68,is also a substrate of PKMTs. Upon oestrogen stimula-tion, ER recruits several co-regulators to the oestrogenresponse elements that modulate activation or repres-sion of target genes. SMYD2 methylates lysine 266 ofER (ERK266) both in cell-free biochemical assays andin breast cancer cells, and this SMYD2-mediated ERmethylation negatively regulates acetylation of lysines266 and 268. Given that acetylation of these lysine resi-dues promotes ER transcriptional activity in responseto the ER ligand 17-oestradiol through enhancing theDNA-binding activity of ER69, SMYD2-dependentlysine 266 methylation attenuates the chromatin recruit-ment of ER to prevent ER target gene activation underan oestrogen-depleted condition69. On oestrogen stim-ulation, ERK266 methylation is diminished, therebyenabling p300/CREB-binding protein (CBP) to acetylateER, which can activate ER target genes. Importantly,ERK266 is also demethylated by LSD1. These resultssuggest a model in which SMYD2 and LSD1 control thedynamics of ERK266 methylation and its crosstalk with

    acetylation of lysines 266 and 268, thereby modulatingER functions in breast cancer cells69(FIG. 4b).

    CCAAT/enhancer-binding protein- (C/EBP) is amember of the basic leucine zipper family of transcriptionregulators that regulates tissue-specific gene expression,proliferation and differentiation in numerous cell types.Increased C/EBP expression has been detected in breastcancer, ovarian tumours and colorectal tumours, andC/EBP is required for tumour progression in the epider-mis70. Pless et al.71demonstrated that lysine 39 of C/EBP(C/EBPK39), which is located in the amino-terminaltransactivation domain, is methylated by EHMT2, leadingto repression of C/EBP transcriptional activity (FIG. 4c).

    REVIEWS

    NATURE REVIEWS |CANCER VOLUME 15 |FEBRUARY 2015 |117

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    9/15

    a b

    cd

    e f

    SETD7

    EHMT2

    SETD6NSD1

    EHMT1

    PHF20

    MeMe

    Me

    Me

    Me Me Me

    LSD1

    LSD1

    PP2A

    PCNA PCNA_N PCNA_C

    SETD8

    StabilizesPCNA protein

    Interaction

    FEN1

    High affinity

    PARP1 DBD

    Automodification domain

    Catalytic domain

    NLS

    SMYD2

    Enhances poly(ADP-ribose)formation aer oxidative stress

    ER

    Transcriptional

    activity

    DBD Hinge Ligand-binding

    Agonistantagonist

    distinction

    K266

    SMYD2

    p300

    K268

    Prevents ERtargetgene activation underan oestrogen-depleted condition

    RELA

    C/EBP

    RHD-n TA1TA2IPT_NF-B

    K218

    K221

    K221

    Me

    K39

    Me

    K248

    Me

    K140

    K310

    Constitutiveactivation ofNF-B activity

    K37

    K180

    Me

    K528

    Gene activation

    in response toTNFstimulation

    FBXL11

    Highaffinity

    Suppression of NF-B-regulated genes throughEHMT1-dependentH3K9 methylation

    STAT3

    K218me1; K221me2

    SETD7

    EZH2

    Promotes STAT3activity in GSCs

    K37me1 K310me1 K266me1; K266me2

    K248me1

    K140me2

    K180me3

    K528me1

    Ac

    DBD LZI II

    TAs

    Repressestranscriptionalactivity

    NTD CC DBD LD

    Negatively regulatestranscription ofsome specific genes

    SH2 CTDY

    Mechanistically, methylation of C/EBP by EHMT2 maycreate a new binding site for a repressive protein com-plex or enhance the interaction between EHMT2 andC/EBP, which promotes H3K9 methylation by EHMT2in the vicinity of C/EBP target genes.

    Signal transducer and activator of transcription 3(STAT3) is a latent transcription factor that acts as anoncoproteinin several cancers72. Intriguingly, STAT3 is

    dimethylated on lysine 140 by SETD7 when it is boundto the subset of promoters that it activates following itstyrosine phosphorylation, and this methylation is revers-ibly demethylated by LSD1 (REF. 73) (FIG. 4d). In this case,STAT3 is methylated in the nucleus and not in the cyto-sol, in particular only when it is part of a promoter-boundcomplex. Moreover, dimethylation only negatively affectssubsets of STAT3-activated genes, implying that lysine

    Figure 4 | Effects of lysine methylation on transcription factors and nuclear proteins. a | Lysine 37 of RELA, a subunit

    of nuclear factor-B (NF-B) , is monomethylated by SETD7, which activates NF-B-dependent genes in response totumour necrosis factor-(TNF) stimulation. Nuclear receptor-binding SET domain-containing protein 1 (NSD1)monomethylates K218 (K218me1) and dimethylates K221 (K221me2), resulting in constitutive NF-B activation throughinteraction with PHF20, which disrupts the recruitment of PP2A to RELA, and these sites are reversibly demethylated by

    Fbox and leucinerich repeat protein 11 (FBXL11). SETD6 monomethylates K310 of RELA and suppresses

    NF-B-regulated genes through euchromatic histone-lysineNmethyltransferase 1 (EHMT1)dependent H3K9methylation. b | SMYD2-mediated oestrogen receptor-(ER) methylation prevents ERtarget gene activation under anoestrogen-depleted condition, and lysine-specific demethylase 1 (LSD1) demethylates this site.c | K39 of CCAAT/

    enhancer-binding protein-(C/EBP) is methylated by EHMT2, which represses C/EBPtranscriptional activity. d | K140 ofsignal transducer and activator of transcription 3 (STAT3) is dimethylated by SETD7, which negatively regulates the

    transcription of various STAT3 target genes. STAT3K140me2 is reversibly demethylated by LSD1. EZH2 trimethylates K180

    of STAT3, which promotes STAT3 activity in glioblastoma stemlike cells (GSCs). e | SETD8 monomethylates K248 of

    proliferating cell nuclear antigen (PCNA), and this methylation stabilizes the PCNA protein and enhances the interaction

    between PCNA and flap endonuclease 1 (FEN1).f| SMYD2 monomethylatesK528 of poly(ADPribose) polymerase 1

    (PARP1) and enhances poly(ADPribose) formation after oxidative stress. CC, coiledcoil domain; CTD, carboxyterminal

    domain; DBD, DNA-binding domain; IPT, immunoglobulin-plexin-transcription; LD, linker domain; LZ, leucine zipperdomain; NLS, nuclear localization signal; NTD, amino terminal domain; PCNA_C, PCNA Cterminal domain; PCNA_N,

    PCNA Nterminal domain; PHF20, PHD finger protein 20; RHD, REL homology domain; SH2, Src homology 2 domain;

    TA, transcriptionactivation domain; Y, conserved tyrosine domain.

    REVIEWS

    118 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    10/15

    Okazaki fragments

    Short, newly synthesized DNA

    fragments that are formed on

    the lagging template strand

    during DNA replication. The

    fragments are produced

    because of the need for DNA

    polymerase to always

    synthesize in a 5-to-3

    direction and are subsequently

    ligated together to form a

    continuous strand.

    methylation may contribute to specific transcriptionalregulation. Furthermore, lysine 180 on STAT3 is also tri-methylated by EZH2. EZH2-dependent STAT3 methyla-tion activates STAT3 functions by increasing its tyrosinephosphorylation in glioblastoma stem-like cells, and thisEZH2STAT3 pathway seems to be one of the key signal-ling nodes in glioblastoma stem-like cells74. Given thatinhibition of EZH2 inhibits the expression of Polycombtarget genes and diminishes STAT3 activity, it might be apotential therapeutic strategy in glioblastoma.

    Other nuclear proteins (PCNA and PARP1).We reportedpreviously that the PKMT SETD8 methylates lysine 248of proliferating cell nuclear antigen (PCNA), one of thekey regulators in DNA replication and cell cycle pro-gression75. Methylation of lysine 248 by SETD8 stabilizesPCNA proteins through inhibition of polyubiquityla-tion and substantially enhances the interaction betweenPCNA and the flap endonuclease FEN1 (REF. 75). Lossof PCNA methylation retards the maturation of Okazakifragmentsand slows DNA replication. Moreover, cells

    expressing a PCNA mutant are unable to be methylatedand are more susceptible to hydrogen peroxide-inducedDNA damage75. There are increased levels of methyl-ated PCNA in cancer cells, and there is a correlationbetween the expression levels of SETD8 and PCNA inhuman cancer tissues75(FIG. 4e). PCNA has been widelyrecognized as a tumour marker for cancer progressionand poor patient prognosis because of its function incell proliferation76. Given this, SETD8-dependent PCNAmethylation is likely to promote tumorigenesis. Hence,inhibition of PCNA methylation by SETD8 might be arational approach to treat cancer.

    Poly(ADP-ribose) polymerase 1 (PARP1) catalyses thetransfer of an ADP ribose unit from its substrate, NAD+,to protein acceptors such as histones and PARP1 itself.The pivotal roles of PARP1 in the DNA repair pathwayprompted researchers to investigate the effect of PARP1inhibition on DNA-damaging anticancer therapies.Indeed, inhibition of PARP1 was shown to enhance thecytotoxicity of DNA-damaging agents to cancer cells77. Weidentified that SMYD2 methylates lysine 528 of PARP1,which is located in the catalytic domain. Lysine 528-meth-ylated PARP1 shows increased poly(ADP-ribosyl)ationactivity after oxidative stress27(FIG. 4f). As SMYD2 deple-tion results in the reduction of PARP1 enzymatic activity,SMYD2 inhibition might increase the susceptibility ofcancer cells to DNA-damaging chemotherapy.

    Somatic mutations of PKMTs and PKDMs

    Many somatic mutations have been discovered inPKMTs and PKDMs through whole-genome sequenc-ing or exome sequencing of cancers (see Supplementaryinformation S1(table)). For example, the missense muta-tions Y641 and A677 in EZH2are frequently observedin diffuse large B cell lymphoma (DLBCL), and thesemutations significantly increase the methyltransferaseactivity of EZH2, resulting in enhanced proliferation ofcancer cells15. In addition, the E1099K missense muta-tion in the PKMT WolfHirschhorn syndrome candi-date 1-like 1 (WHSC1), which is found in 14% of t(12;21)

    ETV6RUNX1-containing acute lymphoblastic leukae-mia cases, enhances the methyltransferase activity ofWHSC1 (REF. 78). It is crucial to consider these tumour-specific mutations for the development of anticancertreatment. Furthermore, somatic mutations in mixed-lineage leukaemia (MLL) lysine methyltransferase fam-ily members and in the PKDM UTX (also known asKDM6A) have frequently been reported in various typesof cancer7990, indicating the importance of these enzymesin human tumorigenesis. So far, the effect of dysregulationof PKMTs and PKDMs caused by somatic mutations hasonly been discussed in the context of their roles in epige-netic regulations through histone methylation or demeth-ylation. Thus, additional studies to clarify non-histonesubstrates and their effects in human cancer are needed.

    Protein lysine methylation as a therapeutic target

    Accumulating evidence clearly indicates that the inhibitionof PKMTs or PKDMs shows promise for the developmentof anticancer therapy. In 2005, Greiner et al.91identifiedthat chaetocin, the first described inhibitor of a PKMT,

    specifically inhibits suppressor of variegation 39 homo-logue 1 (SUV39H1; also known as SU(VAR)3-9) bothin vitroandin vivo.

    Subsequently, BIX-01294, which is an inhibitor of thePKMT EHMT2 (REF. 92), was shown to effectively suppressthe growth of cancer cell lines19(TABLE 2). Analysis of itscrystal structure shows that BIX-01294 lies in a loca-tion occupied by the histone H3 peptide of lysine 4 toarginine 8 (H3K4H3R8) that is next to the methylationsite of lysine 9 (REF. 93), implying that BIX-01294 occu-pies the substrate-binding site. This mode of action ismarkedly different from that of kinase inhibitors becausemost of the anticancer drugs targeting kinases discov-ered so far block phosphotransferase activity by com-peting with ATP94. In 2011, the novel EHMT2 inhibitorUNC0638 with a half-maximal inhibitory concentration(IC

    50) value of

  • 8/9/2019 Nrc 3884Artigo

    11/15

  • 8/9/2019 Nrc 3884Artigo

    12/15

    effectively inhibits the proliferation of EZH2-mutantDLBCL cell lines and xenografts in mice15. The other isEPZ005687, which also targets Y641- and A677-mutatedEZH2 found in non-Hodgkin lymphoma97(TABLE 2).Recently, a new inhibitor (EPZ6438) specific for mutatedEZH2 has been reported that has superior potency andproperties to EPZ005687, including better oral bioavail-ability98(TABLE 2). A Phase I/II clinical trial of EPZ6438has begun for patients with non-Hodgkin lymphoma.By contrast, 10% of patients with myelodysplastic syn-drome (MDS) harbour loss-of-function mutations inEZH2 (REF. 99), and EZH2mutations in these patients areassociated with significantly worse prognosis, althoughthis is not due to transformation to acute myeloid leu-kaemia100. Patients with MDS without monosomy7/del(7q) chromosome anomalies also have reducedexpression of EZH2 in CD34+cells101, which implies thepossibility that EZH2 functions as a tumour suppressorin MDS, and similar results are also reported in myelo-proliferative neoplasm102. Along this line, patients withcolorectal cancer who have EZH2overexpression have a

    good prognosis17. These results highlight that althoughthere is no doubt that it is an ideal therapeutic target,caution is necessary in developing anticancer therapiestargeting EZH2.

    EPZ5676, a potent and selective aminonucleosideinhibitor of the PKMT DOT1L103,104, has also beenstudied in a Phase I clinical trial for the treatment ofpatients with acute leukaemia in which theMLLgenehas undergone rearrangement or tandem duplication(TABLE 2). The data derived from these clinical studiesare expected to provide important information regard-ing further possibilities and potential issues in thedevelopment of anticancer drugs targeting PKMTs.

    With regard to PKDMs as candidates for anticancertherapy development, inhibitors targeting LSD1 havebeen actively studied. LSD1 is significantly overexpressedin human cancer, including small cell lung carcinoma,and has a crucial role in the growth of cancer cellsthrough the regulation of chromatin functions46. Becauseof the similarity in the catalytic and structural proper-ties, drugs targeting monoamine oxidase (MAO) werefirst investigated as LSD1 inhibitors. For example, tranyl-cypromine, an MAO inhibitor used as an antidepressantdrug, can inhibit LSD1 (REF. 105), and tranylcypromineor its analogues show antitumour activity when admin-istered alone or in combination with all-trans-retinoicacid in leukaemia models106. Given that anti-MAO drugs

    are not specific to LSD1 and induce substantial toxicityin vivo, further refined LSD1-specific inhibitors haverecently been developed107. In particular, a Phase I clini-cal trial of GSK2879552, an LSD1-specific inhibitor, hasbeen initiated for patients with relapsed/refractory smallcell lung cancer (TABLE 2). Additionally, a Phase I clinicaltrial of the novel LSD1 inhibitor ORY-1001 has begun foracute myeloid leukaemia (TABLE 2).

    As mentioned above, in addition to the LSD1 fam-ily proteins, JmjC-containing proteins also have PKDMactivity. In 2012, Kruidenier et al.108reported a selectiveJmjC H3K27 demethylase inhibitor. The authors indicatedthe relevance and tractability of demethylase inhibition

    and that targeting the JmjC-containing proteins may havebroad therapeutic application108. Indeed, several JmjC-containing demethylases have been reported as candidatesfor anticancer therapy (TABLE 1), and inhibitors targetingthe Jumonji-type demethylase activity showed antitu-mour effects even though no drugs have yet been evalu-ated in a clinical trial109. Further investigation may reveala great potential of Jumonji-type demethylase inhibitorsas antitumour agents.

    Conclusions and outlook

    Although the first protein lysine methylation was dis-covered in 1959, its physiological significance remainedunknown for a long time. In the twenty-first century, thediscovery of several PKMTs and PKDMs markedly accel-erated a deeper understanding of the role of protein lysinemethylation in many biological processes, particularlyits vital role in epigenetics and also its involvement in

    various human diseases, including cancer. In this regard,although it was difficult to identify methylation sitesof novel substrates, the progress of proteomic analysis

    using mass spectrometry has enabled the identificationof methylation sites to become feasible both in vitroandin vivo. Consequently, lysine methylation is now widelyrecognized as a fundamental PTM. Pang et al.36reported45 high-confidence lysine methylation sites in 40 of 2,607(1.53%) S. cerevisiae proteins, even though their func-tions are mostly unknown. This result demonstrates thepossibility that many substrates of human PKMTs orPKDMs remain uncharacterized. Importantly, severalPKMTs or PKDMs are localized in both the cytoplasmand the nucleus; in particular, SMYD2 and SMYD3 pre-dominantly localize in the cytoplasm. In fact, a couple ofcytoplasmic proteins have been identified as substratesof SMYD2 or SMYD3 (REFS 25,26,33). These facts providehints that important cytoplasmic substrates of PKMTs orPKDMs remain to be identified. Historically, the researchon protein lysine methylation was started to analyse itsepigenetic functions, focusing on histone methylation,and there is no doubt as to its importance for epigeneticregulation. This is also confirmed by the fact that manyresearchers still use the terms histone methyltransferaseand histone demethylase for enzymes relevant to theprotein lysine methylation. Moreover, in addition to itsepigenetic function, further functional analyses mayunveil a wide range of functions of lysine methylationon non-histone proteins and its relevance to humancancer. In this regard, a large number of non-histone

    proteins have been identified as substrates of histoneacetyltransferases and histone deacetylases (HDACs),and many of those are the products of oncogenes ortumour suppressor genes and are directly involved inhuman tumorigenesis110. As HDAC inhibitors are nowconsidered an emerging class of anticancer therapeutics,detailed examinations of the functions of non-histonesubstrates is essential to correctly understand the mecha-nism of actions of HDAC inhibitors110. For the successfuldevelopment of PKMT and PKDM inhibitors for cancertherapy, comprehensive functional analyses to identifycritical substrates of these enzymes in tumorigenesis arealso crucial.

    REVIEWS

    NATURE REVIEWS |CANCER VOLUME 15 |FEBRUARY 2015 |121

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    13/15

    1. Ambler, R. P. & Rees, M. W. -N-methyl-lysine inbacterial flagellar protein. Nature 184, 5657 (1959).

    2. Murray, K. The occurrence of -N-methyl lysine inhistones. Biochemistry 3, 1015 (1964).

    3. Clarke, S. G. & Tamanoi, F. (eds) The Enzymes: Protein

    MethyltransferasesVol. 24(Academic Press, 2006).

    4. Hamamoto, R. et al.SMYD3encodes a histone

    methyltransferase involved in the proliferation of

    cancer cells. Nature Cell Biol. 6, 731740 (2004).

    This is the pioneering report to describe that

    dysregulation of a PKMT can cause tumorigenesis.

    5. Rea, S. et al.Regulation of chromatin structure bysite-specific histone H3 methyltransferases. Nature

    406, 593599 (2000).

    This is the first report to show the existence of a

    PKMT.

    6. Tachibana, M., Sugimoto, K., Fukushima, T. &

    Shinkai, Y. SET domain-containing protein, G9a, is a

    novel lysine-preferring mammalian histone

    methyltransferase with hyperactivity and specific

    selectivity to lysines 9 and 27 of histone H3.J. Biol.

    Chem. 276, 2530925317 (2001).7. Schlichter, A. & Cairns, B. R. Histone trimethylation by

    Set1 is coordinated by the RRM, autoinhibitory, and

    catalytic domains. EMBO J. 24, 12221231 (2005).

    8. Ringrose, L., Ehret, H. & Paro, R. Distinct

    contributions of histone H3 lysine 9 and 27

    methylation to locus-specific stability of polycomb

    complexes. Mol. Cell 16, 641653 (2004).9. Feng, Q. et al.Methylation of H3-lysine 79 is

    mediated by a new family of HMTases without

    a SET domain. Curr. Biol. 12, 10521058 (2002).

    10. Bannister, A. J., Schneider, R. & Kouzarides, T. Histone

    methylation: dynamic or static? Cell 109, 801806

    (2002).

    11. Shi, Y. et al.Histone demethylation mediated by the

    nuclear amine oxidase homolog LSD1. Cell 119,

    941953 (2004).

    This is the first report to show the existence of a

    PKDM.12. Tsukada, Y. et al.Histone demethylation by a family of

    JmjC domain-containing proteins. Nature 439,

    811816 (2006).

    13. Hamamoto, R. et al.Enhanced SMYD3 expression is

    essential for the growth of breast cancer cells. Cancer

    Sci. 97, 113118 (2006).14. Kotake, Y. et al.pRB family proteins are required for

    H3K27 trimethylation and Polycomb repression

    complexes binding to and silencing p16INK4tumorsuppressor gene. Genes Dev. 21, 4954 (2007).

    15. McCabe, M. T. et al.EZH2 inhibition as a therapeutic

    strategy for lymphoma with EZH2-activating

    mutations. Nature 492, 108112 (2012).This study describes the importance of somatic

    mutations in PKMTs in developing anticancer drugs.

    16. Fiskus, W. et al.Highly effective combination of LSD1

    (KDM1A) antagonist and pan-histone deacetylase

    inhibitor against human AML cells. Leukemia28,

    21552164 (2014).

    17. Takawa, M. Validation of the histone methyltransferase

    EZH2 as a therapeutic target for various types of

    human cancer and as a prognostic marker. Cancer Sci.

    102, 12981305 (2011).

    18. Sasaki, M., Yamaguchi, J., Itatsu, K., Ikeda, H. &

    Nakanuma, Y. Over-expression of polycomb group

    protein EZH2 relates to decreased expression of

    p16INK4ain cholangiocarcinogenesis in hepatolithiasis.

    J. Pathol. 215, 175183 (2008).

    19. Cho, H. S. et al.Enhanced expression of EHMT2 is

    involved in the proliferation of cancer cells through

    negative regulation of SIAH1. Neoplasia 13,

    676684 (2011).

    20. Cho, H. S. et al.The JmjC domain-containing histonedemethylase KDM3A is a positive regulator of the

    G1/S transition in cancer cells via transcriptional

    regulation of the HOXA1gene. Int. J. Cancer 131,

    E179189 (2012).21. Toyokawa, G. et al.The histone demethylase JMJD2B

    plays an essential role in human carcinogenesis

    through positive regulation of cyclin-dependent kinase

    6. Cancer Prev. Res. (Phila) 4, 20512061 (2011).

    22. Cho, H. S. et al.RB1 methylation by SMYD2 enhances

    cell cycle progression through an Increase of RB1

    phosphorylation. Neoplasia 14, 476486 (2012).23. Cho, H. S. et al.Enhanced HSP70 lysine methylation

    promotes proliferation of cancer cells through

    activation of Aurora kinase B. Nature Commun. 3,

    1072 (2012).

    This study provides clear evidence that protein

    lysine methylation plays a critical part in the

    regulation of subcellular localization.

    24. Cho, H. S. et al.Demethylation of RB regulator

    MYPT1 by histone demethylase LSD1 promotes cell

    cycle progression in cancer cells. Cancer Res. 71, 16

    (2011).

    25. Hamamoto, R., Toyokawa, G., Nakakido, M., Ueda, K.

    & Nakamura, Y. SMYD2-dependent HSP90

    methylation promotes cancer cell proliferation by

    regulating the chaperone complex formation. Cancer

    Lett.351, 126133 (2014).26. Kunizaki, M. et al.The lysine 831 of vascular

    endothelial growth factor receptor 1 is a novel target

    of methylation by SMYD3. Cancer Res. 67,1075910765 (2007).

    27. Piao, L. et al.The histone methyltransferase SMYD2

    methylates PARP1 and promotes poly(ADP-ribosyl)

    ation activity in cancer cells. Neoplasia 16, 257264.

    e2 (2014).

    28. Smith, B. C. & Denu, J. M. Chemical mechanisms of

    histone lysine and arginine modifications. Biochim.

    Biophys. Acta1789, 4557 (2008).

    29. Shimazu, T., Barjau, J., Sohtome, Y., Sodeoka, M. &

    Shinkai, Y. Selenium-based S-adenosylmethionineanalog reveals the mammalian seven--strandmethyltransferase METTL10 to be an EF1A1 lysine

    methyltransferase. PLoS ONE 9, e105394 (2014).

    30. Fang, R. et al.Human LSD2/KDM1b/AOF1 regulates

    gene transcription by modulating intragenic H3K4me2

    methylation. Mol. Cell 39, 222233 (2010).

    31. Kogure, M. et al.The oncogenic polycomb histone

    methyltransferase EZH2 methylates lysine 120 on

    histone H2B and competes ubiquitination. Neoplasia

    15, 12511261 (2013).

    32. Sone, K. et al.Critical role of lysine 134 methylation

    on histone H2AX for -H2AX production & DNA repair.Nature Commun 5, 5691 (2014).

    33. Mazur, P. K. et al.SMYD3 links lysine methylation of

    MAP3K2 to Ras-driven cancer. Nature 510, 283287

    (2014).

    This study provides strong evidence that a PKMT

    regulates the function of a cytoplasmic protein.34. Khorasanizadeh, S. Recognition of methylated

    histones: new twists and variations. Curr. Opin. Struct.

    Biol. 21, 744749 (2011).

    35. Daze, K. D. & Hof, F. The cation-pi interaction at

    proteinprotein interaction interfaces: developing and

    learning from synthetic mimics of proteins that bind

    methylated lysines.Acc. Chem. Res. 46, 937945

    (2013).

    36. Pang, C. N., Gasteiger, E. & Wilkins, M. R.

    Identification of arginine- and lysine-methylation in the

    proteome of Saccharomyces cerevisiaeand its

    functional implications. BMC Genomics 11, 92 (2010).

    This study comprehensively analyses lysinemethylation of yeast proteins and identifies

    numerous substrates.

    37. Lee, J. M. et al.EZH2 generates a methyl degron

    that is recognized by the DCAF1/DDB1/CUL4 E3

    ubiquitin ligase complex. Mol. Cell 48, 572586

    (2012).

    38. Hodel, M. R., Corbett, A. H. & Hodel, A. E. Dissection

    of a nuclear localization signal.J. Biol. Chem. 276,

    13171325 (2001).

    39. Chuikov, S. et al.Regulation of p53 activity through

    lysine methylation. Nature 432, 353360 (2004).

    This paper provides the first evidence that p53 is a

    substrate of a PKMT.

    40. Stark, G. R., Wang, Y. & Lu, T. Lysine methylation of

    promoter-bound transcription factors and relevance to

    cancer. Cell Res. 21, 375380 (2011).

    41. Lu, T. et al.Regulation of NF-B by NSD1/FBXL11-dependent reversible lysine methylation of

    p65. Proc. Natl Acad. Sci. USA 107, 4651 (2010).

    42. Lu, T. et al.Role of lysine methylation of NF-B indifferential gene regulation. Proc. Natl Acad. Sci. USA

    110, 1351013515 (2013).

    43. Nakamura, Y. Isolation of p53-target genes and their

    functional analysis. Cancer Sci. 95, 711 (2004).

    44. Huang, J. et al.Repression of p53 activity by

    Smyd2-mediated methylation. Nature 444, 629632

    (2006).

    45. Huang, J. et al.p53 is regulated by the lysine

    demethylase LSD1. Nature 449, 105108 (2007).

    This study provides the first evidence that a PKDM

    can also demethylate a non-histone protein.

    46. Hayami, S. et al.Overexpression of LSD1 contributes

    to human carcinogenesis through chromatin

    regulation in various cancers. Int. J. Cancer 128,

    574586 (2011).

    47. Shi, X. et al.Modulation of p53 function by

    SET8-mediated methylation at lysine 382. Mol. Cell

    27, 636646 (2007).

    48. Goodrich, D. W. The retinoblastoma tumor-suppressor

    gene, the exception that proves the rule. Oncogene

    25, 52335243 (2006).

    49. Munger, K. et al.Complex formation of human

    papillomavirus E7 proteins with the retinoblastoma

    tumor suppressor gene product. EMBO J. 8,

    40994105 (1989).

    50. Buchkovich, K., Duffy, L. A. & Harlow, E. The

    retinoblastoma protein is phosphorylated during

    specific phases of the cell cycle. Cell 58, 10971105

    (1989).

    51. Saddic, L. A. et al.Methylation of the retinoblastomatumor suppressor by SMYD2.J. Biol. Chem. 285,

    3773337740 (2010).

    52. Carr, S. M., Munro, S., Kessler, B., Oppermann, U. &

    La Thangue, N. B. Interplay between lysine

    methylation and Cdk phosphorylation in growth

    control by the retinoblastoma protein. EMBO J. 30,

    317327 (2011).

    53. Tsantoulis, P. K. & Gorgoulis, V. G. Involvement of E2F

    transcription factor family in cancer. Eur. J. Cancer 41,

    24032414 (2005).

    54. Chen, H. Z., Tsai, S. Y. & Leone, G. Emerging roles of

    E2Fs in cancer: an exit from cell cycle control. Nature

    Rev. Cancer 9, 785797 (2009).

    55. Kontaki, H. & Talianidis, I. Lysine methylation

    regulates E2F1-induced cell death. Mol. Cell 39,

    152160 (2010).

    56. Ciocca, D. R. & Calderwood, S. K. Heat shock proteins

    in cancer: diagnostic, prognostic, predictive, and

    treatment implications. Cell Stress Chaperones 10,

    86103 (2005).

    57. Trepel, J., Mollapour, M., Giaccone, G. & Neckers, L.

    Targeting the dynamic HSP90 complex in cancer.

    Nature Rev. Cancer 10, 537549 (2010).

    58. Kampinga, H. H. & Craig, E. A. The HSP70 chaperone

    machinery: J proteins as drivers of functional

    specificity. Nature Rev. Mol. Cell Biol. 11, 579592

    (2010).

    59. Wandinger, S. K., Richter, K. & Buchner, J. The Hsp90

    chaperone machinery.J. Biol. Chem. 283,

    1847318477 (2008).

    60. Pratt, W. B. & Toft, D. O. Regulation of signaling

    protein function and trafficking by the hsp90/

    hsp70-based chaperone machinery. Exp. Biol. Med.

    (Maywood) 228, 111133 (2003).

    61. Whitesell, L. & Lindquist, S. L. HSP90 and the

    chaperoning of cancer. Nature Rev. Cancer 5,

    761772 (2005).

    62. Olsson, A. K., Dimberg, A., Kreuger, J. &

    Claesson-Welsh, L. VEGF receptor signalling in

    control of vascular function. Nature Rev. Mol. Cell Biol.

    7, 359371 (2006).63. Fan, F.et al.Expression and function of vascular

    endothelial growth factor receptor-1 on human

    colorectal cancer cells. Oncogene 24, 26472653

    (2005).64. Abdelrahim, M. et al.Regulation of vascular

    endothelial growth factor receptor-1 expression by

    specificity proteins 1, 3, and 4 in pancreatic cancer

    cells. Cancer Res. 67, 32863294 (2007).

    65. Silverman, N. & Maniatis, T. NF-B signaling pathwaysin mammalian and insect innate immunity. Genes Dev.

    15, 23212342 (2001).66. Ea, C. K. & Baltimore, D. Regulation of NF-B activity

    through lysine monomethylation of p65. Proc. Natl

    Acad. Sci. USA 106, 1897218977 (2009).

    67. Levy, D. et al.Lysine methylation of the NF-B subunitRelA by SETD6 couples activity of the histone

    methyltransferase GLP at chromatin to tonic

    repression of NF-B signaling. Nature Immunol. 12,2936 (2011).

    68. Ali, S. & Coombes, R. C. Estrogen receptor in humanbreast cancer: occurrence and significance.

    J. Mammary Gland Biol. Neoplasia 5, 271281

    (2000).

    69. Zhang, X. et al.Regulation of estrogen receptor byhistone methyltransferase SMYD2-mediated protein

    methylation. Proc. Natl Acad. Sci. USA 110,

    1728417289 (2013).

    70. Grimm, S. L. & Rosen, J. M. The role of C/EBPinmammary gland development and breast cancer.

    J. Mammary Gland Biol. Neoplasia 8, 191204

    (2003).

    71. Pless, O. et al.G9a-mediated lysine methylation

    alters the function of CCAAT/enhancer-binding

    protein-.J. Biol. Chem. 283, 2635726363(2008).

    72. Chen, Y. et al.STAT3, a poor survival predicator, is

    associated with lymph node metastasis from breast

    cancer.J. Breast Cancer 16, 4049 (2013).

    REVIEWS

    122 |FEBRUARY 2015 |VOLUME 15 www.nature.com/reviews/cancer

    2015 Macmillan Publishers Limited. All rights reserved

  • 8/9/2019 Nrc 3884Artigo

    14/15

    73. Yang, J. et al.Reversible methylation of promoter-

    bound STAT3 by histone-modifying enzymes. Proc.

    Natl Acad. Sci. USA 107, 2149921504 (2010).

    74. Kim, E. et al.Phosphorylation of EZH2 activates

    STAT3 signaling via STAT3 methylation and promotes

    tumorigenicity of glioblastoma stem-like cells. Cancer

    Cell 23, 839852 (2013).

    This study provides clear evidence that non-histone

    lysine methylation is an important target of cancer

    therapy in vivo.

    75. Takawa, M. et al.Histone lysine methyltransferase

    SETD8 promotes carcinogenesis by deregulatingPCNA expression. Cancer Res. 72, 32173227

    (2012).

    76. Zhao, H. et al.Targeting tyrosine phosphorylation of

    PCNA inhibits prostate cancer growth. Mol. Cancer

    Ther. 10, 2936 (2011).

    77. Chen, A. PARP inhibitors: its role in treatment of

    cancer. Chin. J. Cancer 30, 463471 (2011).

    78. Jaffe, J. D. et al.Global chromatin profiling reveals

    NSD2 mutations in pediatric acute lymphoblastic

    leukemia. Nature Genet. 45, 13861391 (2013).

    This study describes the importance of somatic

    mutations in PKMTs for developing anticancer

    drugs.

    79. Yin, S. et al.Exome sequencing identifies frequent

    mutation of MLL2in non-small cell lung carcinoma

    from Chinese patients. Sci. Rep. 4, 6036 (2014).

    80. Gossage, L. et al.Clinical and pathological impact of

    VHL, PBRM1, BAP1, SETD2, KDM6A, and JARID1c in

    clear cell renal cell carcinoma. Genes Chromosomes

    Cancer 53, 3851 (2014).

    81. Cleary, S. P.et al.Identification of driver genes in

    hepatocellular carcinoma by exome sequencing.

    Hepatology 58, 16931702 (2013).

    82. Dubuc, A. M. et al.Aberrant patterns of H3K4 and

    H3K27 histone lysine methylation occur across

    subgroups in medulloblastoma.Acta Neuropathol.

    125, 373384 (2013).

    83. Jankowska, A. M. et al.Mutational spectrum analysis

    of chronic myelomonocytic leukemia includes genes

    associated with epigenetic regulation: UTX, EZH2, and

    DNMT3A. Blood 118, 39323941 (2011).

    84. Morin, R. D. Frequent mutation of histone-modifying

    genes in non-Hodgkin lymphoma. Nature 476,

    298303 (2011).

    85. Wang, X. X. et al.Somatic mutations of the mixed-

    lineage leukemia 3 (MLL3) gene in primary breast

    cancers. Pathol. Oncol. Res. 17, 429433 (2011).

    86. Gui, Y. Frequent mutations of chromatin remodeling

    genes in transitional cell carcinoma of the bladder.

    Nature Genet. 43, 875878 (2011).

    87. Grasso, C. S. et al.The mutational landscape of lethalcastration-resistant prostate cancer. Nature 487,

    239243 (2012).

    88. Lindberg, J. et al.The mitochondrial and autosomal

    mutation landscapes of prostate cancer. Eur. Urol. 63,

    702708 (2013).

    89. Ho, A. S. et al.The mutational landscape of adenoid

    cystic carcinoma. Nature Genet. 45, 791798 (2013).

    90. Kandoth, C. et al.Mutational landscape and

    significance across 12 major cancer types. Nature

    502, 333339 (2013).

    91. Greiner, D., Bonaldi, T., Eskeland, R., Roemer, E. &

    Imhof, A. Identification of a specific inhibitor of the

    histone methyltransferase SU(VAR)3-9. Nature Chem.

    Biol. 1, 143145 (2005).

    92. Kubicek, S. et al.Reversal of H3K9me2 by a small-

    molecule inhibitor for the G9a histone

    methyltransferase. Mol. Cell 25, 473481 (2007).

    93. Chang, Y. et al.Structural basis for G9a-like protein

    lysine methyltransferase inhibition by BIX-01294.

    Nature Struct. Mol. Biol. 16, 312317 (2009).This study determined the crystal structure of the

    catalytic SET domain of G9a-like protein in complex

    with the specific inhibitor BIX-01294.

    94. Liu, Y. & Gray, N. S. Rational design of inhibitors that

    bind to inactive kinase conformations. Nature Chem.

    Biol. 2, 358364 (2006).

    95. Vedadi, M. et al.A chemical probe selectively inhibits

    G9a and GLP methyltransferase activity in cells.

    Nature Chem. Biol. 7, 566574 (2011).

    96. Ferguson, A. D. et al.Structural basis of substrate

    methylation and inhibition of SMYD2. Structure 19,

    12621273 (2011).

    This study describes a drug development strategy

    for inhibitors of a PKMT using a non-histone

    protein as a substrate.

    97. Knutson, S. K. et al.A selective inhibitor of EZH2

    blocks H3K27 methylation and kills mutant lymphoma

    cells. Nature Chem. Biol. 8, 890896 (2012).

    98. Knutson, S. K. et al.Selective inhibition of EZH2 by

    EPZ-6438 leads to potent antitumor activity in

    EZH2-mutant non-Hodgkin lymphoma. Mol. Cancer

    Ther. 13, 842854 (2014).

    99. Nikoloski, G. et al.Somatic mutations of the histone

    methyltransferase gene EZH2in myelodysplastic

    syndromes. Nature Genet. 42, 665667 (2010).

    100. Bejar, R. et al.Validation of a prognostic model and

    the impact of mutations in patients with lower-risk

    myelodysplastic syndromes.J. Clin. Oncol. 30,

    33763382 (2012).

    101. Jerez, A. et al.Loss of heterozygosity in 7q myeloiddisorders: clinical associations and genomic

    pathogenesis. Blood 119, 61096117 (2012).

    102. Muto, T. et al.Concurrent loss of Ezh2 and Tet2

    cooperates in the pathogenesis of myelodysplastic

    disorders.J. Exp. Med. 210, 26272639 (2013).

    103. Basavapathruni, A. et al.Nonclinical pharmacokinetics

    and metabolism of EPZ-5676, a novel DOT1L histone

    methyltransferase inhibitor. Biopharm. Drug Dispos.

    35, 237252 (2014).

    104. Klaus, C. R. et al.DOT1L inhibitor EPZ-5676 displays

    synergistic antiproliferative activity in combination

    with standard of care drugs and hypomethylating

    agents in MLL-rearranged leukemia cells.

    J. Pharmacol. Exp. Ther. 350, 646656 (2014).

    References 103 and 104 describe the DOT1L

    inhibitor EPZ5676, which is currently in clinical

    trials.

    105. Schmidt, D. M. & McCafferty, D. G.

    Trans-2-phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1.

    Biochemistry 46, 44084416 (2007).

    106. Schenk, T. et al.Inhibition of the LSD1 (KDM1A)

    demethylase reactivates the all-trans-retinoic acid

    differentiation pathway in acute myeloid leukemia.

    Nature Med. 18, 605611 (2012).

    107. Wang, J. et al.Novel histone demethylase LSD1

    inhibitors selectively target cancer cells with

    pluripotent stem cell properties. Cancer Res. 71,

    72387249 (2011).

    108. Kruidenier, L. et al.A selective jumonji H3K27

    demethylase inhibitor modulates the proinflammatory

    macrophage response. Nature 488, 404408

    (2012).

    This report provides evidence that a Jumonji-type

    demethylase is a good target for drug discovery.109. Wang, L. et al.A small molecule modulates Jumonji

    histone demethylase activity and selectively inhibits

    cancer growth. Nature Commun. 4, 2035 (2013).

    110. Singh, B. N. et al.Nonhistone protein acetylation as

    cancer therapy targets. Expert Rev. Anticancer Ther.

    10, 935954 (2010).111. Komatsu, S. et al.Overexpression of SMYD2 relates

    to tumor cell proliferation and malignant outcome of

    esophageal squamous cell carcinoma. Carcinogenesis

    30, 11391146 (2009).

    112. Abu-Farha, M. et al.The tale of two domains:

    proteomics and genomics analysis of SMYD2, a new

    histone methyltransferase. Mol. Cell Proteom. 7,

    560572 (2008).

    113. Brown, M. A., Sims, R. J., 3rd, Gottlieb, P. D. &

    Tucker, P. W. Identification and characterization of

    Smyd2: a split SET/MYND domain-containing histone

    H3 lysine 36-specific methyltransferase that interacts

    with the Sin3 histone deacetylase complex. Mol.

    Cancer 5, 26 (2006).

    114. Silva, F. P. et al.Enhanced methyltransferase activity

    of SMYD3 by the cleavage of its N-terminal region in

    human cancer cells. Oncogene 27, 26862692

    (2008).115. Foreman, K. W. et al.Structural and functional

    profiling of the human histone methyltransferaseSMYD3. PLoS ONE 6, e22290 (2011).

    116. Dong, S. W.et al.Effect of the downregulation of

    SMYD3 expression by RNAi on RIZ1 expression and

    proliferation of esophageal squamous cell carcinoma.

    Oncol. Rep. 32,10641070 (2014).117. Sponziello, M. et al.Epigenetic-related gene

    expression profile in medullary thyroid cancer revealed

    the overexpression of the histone methyltransferases

    EZH2 and SMYD3 in aggressive tumours. Mol. Cell

    Endocrinol. 392, 813 (2014).

    118. Vieira, F. Q. et al.Deregulated expression of

    selected histone methylases and demethylases in

    prostate carcinoma. Endocr. Relat. Cancer 21,

    5161 (2014).

    119. Wang, S. Z. et al.Knockdown of SMYD3 by RNA

    interference inhibits cervical carcinoma cell growth

    and invasion in vitro. BMB Rep. 41, 294299

    (2008).

    120. Zeng, B. et al.Epigenetic regulation of miR-124 by

    hepatitis C virus core protein promotes migration and

    invasion of intrahepatic cholangiocarcinoma cells by

    targeting SMYD3. FEBS Lett. 586, 32713278

    (2012).

    121. Guo, N. et al.Hepatitis C virus core upregulates the

    methylation status of the RASSF1Apromoter through

    regulation of SMYD3 in hilar cholangiocarcinoma cells.

    Acta Biochim. Biophys. Sin. (Shanghai) 43, 354361

    (2011).

    122. Wang, G. G., Cai, L., Pasillas, M. P. & Kamps, M. P.

    NUP98NSD1 links H3K36 methylation to Hox-Agene activation and leukaemogenesis. Nature Cell

    Biol. 9, 804812 (2007).

    123. Panarello, C., Rosanda, C. & Morerio, C. Cryptic

    translocation t(5;11)(q35;p15.5) with involvement of

    the NSD1and NUP98genes without 5q deletion in

    childhood acute myeloid leukemia. Genes

    Chromosomes Cancer 35, 277281 (2002).

    124. Hollink, I. H. et al.NUP98/NSD1 characterizes a novel

    poor prognostic group in acute myeloid leukemia with

    a distinct HOXgene expression pattern. Blood 118,

    36453656 (2011).

    125. Toyokawa, G. et al.Histone lysine methyltransferase

    Wolf-Hirschhorn syndrome candidate 1 is involved

    in human carcinogenesis through regulation of

    the Wnt pathway. Neoplasia 13, 887898

    (2011).

    126. Martinez-Garcia, E. et al.The MMSET histone methyl

    transferase switches global histone methylation and

    alters gene expression in t(4;14) multiple myeloma

    cells. Blood 117, 211220 (2011).

    127. Ezponda, T. et al.The histone methyltransferase

    MMSET/WHSC1 activates TWIST1 to promote an

    epithelial-mesenchymal transition and invasive

    properties of prostate cancer. Oncogene 32,

    28822890 (2013).

    128. Kim, J. Y. et al.Multiple-myeloma-related WHSC1/

    MMSET isoform RE-IIBP is a histone

    methyltransferase with transcriptional repression

    activity. Mol. Cell. Biol. 28, 20232034 (2008).

    129. Pei, H. et al.MMSET regulates histone H4K20

    methylation and 53BP1 accumulation at DNA

    damage sites. Nature 470, 124128 (2011).

    130. Kang, D. et al.The histone methyltransferase

    WolfHirschhorn syndrome candidate 1-like 1

    (WHSC1L1) is involved in human carcinogenesis.

    Genes Chromosomes Cancer 52, 126139

    (2013).

    131. Zhou, Z., Thomsen, R., Kahns, S. & Nielsen, A. L.

    The NSD3L histone methyltransferase regulates cell

    cycle and cell invasion in breast cancer cells.

    Biochem. Biophys. Res. Commun. 398, 565570(2010).

    132. Kim, S. M. et al.Characterization of a novel

    WHSC1-associated SET domain protein with H3K4

    and H3K27 methyltransferase activity. Biochem.

    Biophys. Res. Commun. 345, 318323 (2006).

    133. Salz, T.et al.hSETD1A regulates Wnt target genes

    and controls tumor growth of colorectal cancer cells.

    Cancer Res. 74, 775786 (2014).

    134. Hu, H. Y.et al.Set9, NF-B, and microRNA-21mediate berberine-induced apoptosis of human

    multiple myeloma cells.Acta Pharmacol. Sin.34,

    157166 (2013).

    135. Subramanian, K. et al.Regulation of estrogen receptor

    by the SET7 lysine methyltransferase. Mol. Cell 30,336347 (2008).

    136. OCarroll, D. et al.Isolation and characterization of

    Suv39h2, a second histone H3 methyltransferase

    gene that displays testis-specific expression. Mol. Cell.

    Biol. 20, 94239433 (2000).

    137. Lehnertz, B. et al.The methyltransferase G9aregulates HoxA9-dependent transcription in AML.

    Genes Dev. 28, 317327 (2014).

    138. Zhong, X. et al.Overexpression of G9a and MCM7 in

    esophageal squamous cell carcinoma associated with

    poor prognosis. Histopathologyhttp://dx.doi.

    org/10.1111/his.12456(2014).

    139. Natarajan, T. G. et al.Epigenetic regulator MLL2

    shows altered expression in cancer cell lines and

    tumors from human breast and colon. Cancer Cell. Int.

    10, 13 (2010).

    140. Xia, M. et al.Downregulation of MLL3 in

    esophageal squamous cell carcinoma is required for

    the growth and metastasis of cancer cells. Tumour

    Biol.http://dx.doi.org/10.1007/s13277-014-2616-3

    (2014).141. Li, B. et al.Association of MLL3 expression with

    prognosis in gastric cancer. Genet. Mol. Res. 13,

    75137518 (2014).

    REVIEWS

    NATURE REVIEWS |CANCER VOLUME 15 |FEBRUARY 2015 |123

    2015 Macmillan Publishers Limited. All rights reserved

    http://dx.doi.org/10.1111/his.12456http://dx.doi.org/10.1111/his.12456http://dx.doi.org/10.1111/his.12456http://dx.doi.org/10.1007/s13277-014-2616-3http://dx.doi.org/10.1007/s13277-014-2616-3http://dx.doi.org/10.1111/his.12456http://dx.doi.org/10.1111/his.12456
  • 8/9/2019 Nrc 3884Artigo

    15/15

    142. Ruault, M., Brun, M. E., Ventura, M., Roizes, G. & De

    Sario, A. MLL3, a new human member of the TRX/

    MLL gene family, maps to 7q36, a chromosome region

    frequently deleted in myeloid leukaemia. Gene 284,

    7381 (2002).

    143. Okada, Y. et al.hDOT1L links histone methylation to

    leukemogenesis. Cell 121, 167178 (2005).

    144. Nguyen, A. T., Taranova, O., He, J. & Zhang, Y. DOT1L,

    the H3K79 methyltransferase, is required for

    MLL-AF9-mediated leukemogenesis. Blood 117,

    69126922 (2011).

    145. Bernt, K. M. & Armstrong, S. A. A role for DOT1L inMLL-rearranged leukemias. Epigenomics 3, 667670

    (2011).

    146. Yamada, D. et al.Role of the hypoxia-related gene,

    JMJD1A, in hepatocellular carcinoma: clinical impact

    on recurrence after hepatic resection.Ann. Surg.

    Oncol. 19, S355S364 (2012).

    147. Guo, X. et al.The expression of histone demethylase

    JMJD1Ain renal cell carcinoma. Neoplasma 58,

    153157 (2011).

    148. Kogure, M. et al.Deregulation of the histone

    demethylase JMJD2A is involved in human

    carcinogenesis through regulation of the G1/S

    transition. Cancer Lett.336, 7684 (2013).

    149. Berry, W. L., Shin, S., Lightfoot, S. A. & Janknecht, R.

    Oncogenic features of the JMJD2A histone

    demethylase in breast cancer. Int. J. Oncol. 41,

    17011706 (2012).

    150. Wang, H. L. et al.Expression and effects of JMJD2A

    histone demethylase in endometrial carcinoma.

    Asian Pac. J. Cancer Prev. 15, 30513056 (2014).

    151. Hu, C. E., Liu, Y. C., Zhang, H. D. & Huang, G. J.

    JMJD2A predicts prognosis and regulates cell growth

    in human gastric cancer. Biochem. Biophys. Res.

    Commun. 449, 17 (2014).152. Sun, B. B. et al.Silencing of JMJD2B induces cell

    apoptosis via mitochondria-mediated and death

    receptor-mediated pathway activation in colorectal

    cancer.J. Dig. Dis.15, 491500 (2014).

    153. Chen, L. et al.Jumonji domain-containing protein 2B

    silencing induces DNA damage response via STAT3

    pathway in colorectal cancer. Br. J. Cancer 110,

    10141026 (2014).

    154. Zhao, L. et al.JMJD2B promotes epithelial-

    mesenchymal transition by cooperating with -cateninand enhances gastric cancer metastasis. Clin. CancerRes. 19, 64196429 (2013).

    155. Ene, C. I. et al.Histone demethylase Jumonji D3

    (JMJD3) as a tumor suppressor by regulating p53

    protein nuclear stabilization. PLoS ONE 7, e51407

    (2012).

    156. Nickerson, M. L. et al.Concurrent alterations in

    TERT, KDM6A, and the BRCA pathway in

    bladder cancer. Clin. Cancer Res. 20,

    49354948 (2014).

    157. Ntziachristos, P. et al.Contrasting roles of histone 3

    lysine 27 demethylases in acute lymphoblastic

    leukaemia. Nature514, 513517 (2014).

    158. Shen, Y. et al.Expression and significance of histone

    H3K27 demethylases in renal cell carcinoma. BMC

    Cancer 12, 470 (2012).

    159. van Haaften, G. et al.Somatic mutations of the histone

    H3K27 demethylase gene UTXin human cancer.

    Nature Genet. 41, 521523 (2009).

    160. Hayami, S. et al.Overexpression of the JmjC histone

    demethylase KDM5B in human carcinogenesis:

    involvement in the proliferation of cancer cells

    through the E2F/RB pathway. Mol. Cancer 9, 59

    (2010).161. Yamamoto, S. et al.JARID1Bis a luminal lineage-

    driving oncogene in breast cancer. Cancer Cell 25,

    762777 (2014).

    162. Kano, Y. et al.Jumonji/Arid1b (Jarid1b) protein

    modulates human esophageal cancer cell growth. Mol.

    Clin. Oncol. 1, 753757 (2013).

    163. Ohta, K. et al.Depletion of JARID1B induces cellular

    senescence in human colorectal cancer. Int. J. Oncol.

    42, 12121218 (2013).

    164. Radberger, P., Radberger, A., Bykov, V. J.,Seregard, S. & Economou, M. A. JARID1B protein

    expression and prognostic implications in uveal

    melanoma. Invest. Ophthalmol. Vis. Sci. 53,

    44424449 (2012).

    AcknowledgementsThe authors express great gratitude to the past and present

    members of Y.N.s laboratory.

    Competing interests statementThe authors declare competing interests: see Web version for

    details.

    DATABASESNational Cancer Institute Drug Dictionary:

    http://www.cancer.gov/drugdictionary

    Pathway Interaction Database:http://pid.nci.nih.gov

    SUPPLEMENTARY INFORMATIONSee online article:S1(table)

    ALL LINKS ARE ACTIVE IN THE ONLINE PDF

    REVIEWS

    http://www.nature.com/nrc/journal/v15/n2/full/nrc3884.html#affil-authhttp://www.cancer.gov/drugdictionaryhttp://pid.nci.nih.gov/http://pid.nci.nih.gov/http://www.nature.com/nrc/journal/v15/n2/full/nrc3884.html#supplementary-informationhttp://www.nature.com/nrc/journal/v15/n2/full/nrc3884.html#supplementary-informationhttp://www.nature.com/nrc/journal/v15/n2/full/nrc3884.html#supplementary-informationhttp://pid.nci.nih.gov/http://www.cancer.gov/drugdictionaryhttp://www.nature.com/nrc/journal/v15/n2/full/nrc3884.html#affil-auth