Cancer Letters

Cancer Letters

Volume 458, 28 August 2019, Pages 56-65
Cancer Letters

Mini-review
COMPASS Ascending: Emerging clues regarding the roles of MLL3/KMT2C and MLL2/KMT2D proteins in cancer

https://doi.org/10.1016/j.canlet.2019.05.024Get rights and content

Highlights

  • KMT2C/D methyltransferases are frequently mutated in cancer.

  • KMT2C/D mutations frequently co-occur in lung cancer.

  • Somatic loss of KMT2C/D function as drivers of oncogenesis.

  • KMT2C/D function to maintain epithelial states.

  • MLR COMPASS complexes control gene transcriptional enhancer functions.

Abstract

The KMT2 (lysine methyltransferase) family of histone modifying proteins play essential roles in regulating developmental pathways, and mutations in the genes encoding these proteins have been strongly linked to many blood and solid tumor cancers. The KMT2A-D proteins are histone 3 lysine 4 (H3K4) methyltransferases embedded in large COMPASS-like complexes important for RNA Polymerase II-dependent transcription. KMT2 mutations were initially associated with pediatric Mixed Lineage Leukemias (MLL) and found to be the result of rearrangements of the MLL1/KMT2A gene at 11q23. Over the past several years, large-scale tumor DNA sequencing studies have revealed the potential involvement of other KMT2 family genes, including heterozygous somatic mutations in the paralogous MLL3/KMT2C and MLL2(4)/KMT2D genes that are now among the most frequently associated with human cancer. Recent studies have provided a better understanding of the potential roles of disrupted KMT2C and KMT2D family proteins in cell growth aberrancy. These findings, together with an examination of cancer genomics databases provide new insights into the contribution of KMT2C/D proteins in epigenetic gene regulation and links to carcinogenesis.

Introduction

Post-translational histone modifications provide a vast epigenetic regulatory language that can be written and decoded by a large number of proteins and are important for controlling the timing and level of gene expression among all eucaryotes [1]. The methylation of lysine residues on N-terminal histone tails can provide both gene activation and repression signals and is one of the best-studied epigenetic modifications [2]. Methylation of histone 3 lysine 4 (H3K4) is frequently associated with active transcription, with di- and trimethylated H3K4 associated with active gene promoters and monomethylation found at gene enhancers [3]. The enzymes that place methyl modifications or marks on H3K4 are known as the KMT2 (lysine methyltransferase) family and are found embedded within enormous protein complexes, named COMPASS (COMplex of Proteins ASsociated with SET1) [4]. The discovery almost 30 years ago of recurrent translocation-associated leukemias involving MLL1/KMT2A, a homolog of the Drosophila Trithorax (Trx) protein important in Hox gene regulation, provided a critical platform for investigations into the roles of the KMT2 proteins in animal development and cancer [[5], [6], [7]]. More recently, genes encoding other KMT2 family proteins have been implicated in cancer [8]. Tumor genome and exome sequencing studies combined with cancer cell and model system genetic analyses over the past decade have revealed an unanticipated vital role for the KMT2 family enzymes in a diverse set of cancers, both as drivers of oncogenesis and as critical cooperating mutations in both cancer progression and post-therapy relapse.

Section snippets

KMT2 COMPASS-like complexes

KMT2 family proteins (SET1A, SET1B, MLL1/KMT2A, MLL4(2)/KMT2B, MLL3/KMT2C and MLL2(4)/KMT2D) provide the histone lysine methyltransferase activity of mammalian COMPASS complexes. Designated MLL1-5 based on relatedness to the MLL1 gene (Mixed Lineage Leukemia), KMT2 proteins comprise three subgroups, called COMPASS and COMPASS-like based homology with Drosophila Trx, Trr and dSet1 [4,8]. Genome duplication during mammalian evolution resulted in two paralogs in each KMT2 subgroup. KMT2A/MLL1 and

Emerging roles for KMT2C and KMT2D in cancer

The epigenetic functions of the COMPASS complexes are vital for normal animal development and frequent mutations affecting these proteins or other components of the COMPASS complexes are associated with a large number of cancers [32]. Mutations involving KMT2 family genes are among the most commonly seen in many diverse cancer types and there is a statistically significant co-occurrence, raising the possibility that the simultaneous disabling of several KMT2 genes might be a critical step in

Lessons from KMT2C/D mutations in lung cancer

Lung cancers, both non small cell (NSCLC, 85%) and small cell (SCLC, 15%), are a leading cause of cancer-associated deaths worldwide with few treatment options and a high recurrence rate. SCLC is a highly aggressive neuroendocrine tumor with a very poor prognosis in which TP53 and RB1 are frequently mutated. Several whole genome or exome sequencing studies have also revealed a high frequency of KMT2D (8–24%) but not KMT2C mutations [[55], [56], [57], [58], [59], [60], [61]]. These studies

Functional distinctions between KMT2C and KMT2D in cancer

Possible oncogenic mechanisms associated with KMT2C/D loss involve the dysregulation of transcriptional enhancers and transcription factor-dependent programs that drive cellular pathways required for tumor suppression and anti-tumor immune evasion [12,21]. For example, several studies have demonstrated important direct interactions between KMT2C/D and the tumor suppressor TP53 [[74], [75], [76], [77], [78]]. The activation of TP53 target genes following doxorubicin (DNA damaging agent)

Germline roles of KMT2-family genes in cancer predisposition

The KMT2A-D genes are each essential for organismal viability, with strong depletion, homozygous loss of function mutations or deletions resulting in embryonic or perinatal lethality (KMT2A/MLL1: [86,87]; KMT2B/MLL4(2): [88]; KMT2C/MLL3 and KMT2D/MLL2(4): [81,89]. Somatic mutations in the KMT2A-D genes have been strongly linked to cancer development; however, it is less clear the impacts of de novo germline mutations in cancer predisposition. The KMT2A-D genes have each been associated with

Conclusions and perspectives

Cancer exome databases reveal that the KMT2C/MLL3 and KMT2D/MLL2(4) histone lysine methyltransferases are among the most frequently mutated genes across a variety of cancer types. Most of these cancers are associated with mutations that alter the proteins through missense changes or truncations resulting in reduced functions, supporting their roles as tumor suppressors. Genetic removal of these genes in cancer cells, embryonic stem cells, as well as both vertebrate and invertebrate animal

Author contributions

AD and RF conceived the idea for the review, researched the literature and wrote the manuscript.

Conflict of interest statement

The authors declare that they have no competing financial interests.

Acknowledgements

We thank Claudia Zraly, David Ford and Matthew Kroll for helpful advice and comments on the manuscript. R. F. was supported by a NIH T35 award from the NHLBI (HL120835). The research in the laboratory of A.D. is supported by the National Science Foundation (MCB1716431).

References (104)

  • S. Umemura et al.

    Therapeutic priority of the PI3K/AKT/mTOR pathway in small cell lung cancers as revealed by a comprehensive genomic analysis

    J. Thorac. Oncol.

    (2014)
  • T. Vavala et al.

    Precision medicine in age-specific non-small-cell-lung-cancer patients: integrating biomolecular results into clinical practice-A new approach to improve personalized translational research

    Lung Cancer

    (2017)
  • M. Imielinski et al.

    Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing

    Cell

    (2012)
  • M. Choi et al.

    Mutation profiles in early-stage lung squamous cell carcinoma with clinical follow-up and correlation with markers of immune function

    Ann. Oncol.

    (2017)
  • W. Xiong et al.

    Downregulation of KMT2D suppresses proliferation and induces apoptosis of gastric cancer

    Biochem. Biophys. Res. Commun.

    (2018)
  • H. Rahnamoun et al.

    Mutant p53 regulates enhancer-associated H3K4 monomethylation through interactions with the methyltransferase MLL4

    J. Biol. Chem.

    (2018)
  • W.D. Jones et al.

    De novo mutations in MLL cause Wiedemann-Steiner syndrome

    Am. J. Hum. Genet.

    (2012)
  • H. Zhang et al.

    A novel deletion mutation in KMT2A identified in a child with ID/DD and blood eosinophilia

    BMC Med. Genet.

    (2019)
  • M. Zech et al.

    Haploinsufficiency of KMT2B, encoding the lysine-specific histone methyltransferase 2B, results in early-onset generalized dystonia

    Am. J. Hum. Genet.

    (2016)
  • K.M. Gorman et al.

    Review of the phenotype of early-onset generalised progressive dystonia due to mutations in KMT2B

    Eur. J. Paediatr. Neurol.

    (2018)
  • T. Kleefstra et al.

    Disruption of an EHMT1-associated chromatin-modification module causes intellectual disability

    Am. J. Hum. Genet.

    (2012)
  • E.L. Greer et al.

    Histone methylation: a dynamic mark in health, disease and inheritance

    Nat. Rev. Genet.

    (2012)
  • A. Shilatifard

    The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis

    Annu. Rev. Biochem.

    (2012)
  • R.C. Rao et al.

    Hijacked in cancer: the KMT2 (MLL) family of methyltransferases

    Nat. Rev. Canc.

    (2015)
  • R.K. Slany

    When epigenetics kills: MLL fusion proteins in leukemia

    Hematol. Oncol.

    (2005)
  • R.K. Slany

    The molecular mechanics of mixed lineage leukemia

    Oncogene

    (2016)
  • M. Mohan et al.

    The COMPASS family of H3K4 methylases in Drosophila

    Mol. Cell. Biol.

    (2011)
  • D. Hu et al.

    The MLL3/MLL4 branches of the COMPASS family function as major histone H3K4 monomethylases at enhancers

    Mol. Cell. Biol.

    (2013)
  • A. Piunti et al.

    Epigenetic balance of gene expression by Polycomb and COMPASS families

    Science

    (2016)
  • C. Wang et al.

    Enhancer priming by H3K4 methyltransferase MLL4 controls cell fate transition

    Proc. Natl. Acad. Sci. U. S. A.

    (2016)
  • B. Lai et al.

    MLL3/MLL4 are required for CBP/p300 binding on enhancers and super-enhancer formation in brown adipogenesis

    Nucleic Acids Res.

    (2017)
  • S.P. Wang et al.

    A UTX-MLL4-p300 transcriptional regulatory network coordinately shapes active enhancer landscapes for eliciting transcription

    Mol. Cell

    (2017)
  • F. Tie et al.

    Histone demethylase UTX and chromatin remodeler BRM bind directly to CBP and modulate acetylation of histone H3 lysine 27

    Mol. Cell. Biol.

    (2012)
  • R. Baas et al.

    The mixed lineage leukemia 4 (MLL4) methyltransferase complex is involved in transforming growth factor beta (TGF-beta)-activated gene transcription

    Transcription

    (2018)
  • A. Ortega-Molina et al.

    The histone lysine methyltransferase KMT2D sustains a gene expression program that represses B cell lymphoma development

    Nat. Med.

    (2015)
  • J.K. Rhee et al.

    Identification of local clusters of mutation hotspots in cancer-related genes and their biological relevance

    IEEE ACM Trans. Comput. Biol. Bioinform

    (2018)
  • C. Qian et al.

    SET domain protein lysine methyltransferases: structure, specificity and catalysis

    Cell. Mol. Life Sci.

    (2006)
  • K.M. Dorighi et al.

    Mll3 and Mll4 facilitate enhancer RNA synthesis and transcription from promoters independently of H3K4 monomethylation

    Mol. Cell

    (2017)
  • R. Rickels et al.

    Histone H3K4 monomethylation catalyzed by Trr and mammalian COMPASS-like proteins at enhancers is dispensable for development and viability

    Nat. Genet.

    (2017)
  • S. Weirich et al.

    Somatic cancer mutations in the MLL1 histone methyltransferase modulate its enzymatic activity and dependence on the WDR5/RBBP5/ASH2L complex

    Mol Oncol

    (2017)
  • S. Weirich et al.

    Somatic cancer mutations in the MLL3-SET domain alter the catalytic properties of the enzyme

    Clin. Epigenet.

    (2015)
  • N.J. Zeleznik-Le et al.

    11q23 translocations split the "AT-hook" cruciform DNA-binding region and the transcriptional repression domain from the activation domain of the mixed-lineage leukemia (MLL) gene

    Proc. Natl. Acad. Sci. U. S. A.

    (1994)
  • R.K. Slany et al.

    The oncogenic capacity of HRX-ENL requires the transcriptional transactivation activity of ENL and the DNA binding motifs of HRX

    Mol. Cell. Biol.

    (1998)
  • C. Bach et al.

    Alterations of the CxxC domain preclude oncogenic activation of mixed-lineage leukemia 2

    Oncogene

    (2009)
  • Z.B. Xia et al.

    MLL repression domain interacts with histone deacetylases, the polycomb group proteins HPC2 and BMI-1, and the corepressor C-terminal-binding protein

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • Y. Sedkov et al.

    Methylation at lysine 4 of histone H3 in ecdysone-dependent development of Drosophila

    Nature

    (2003)
  • D. Fantini et al.

    A Carcinogen-induced mouse model recapitulates the molecular alterations of human muscle invasive bladder cancer

    Oncogene

    (2018)
  • W. Yang et al.

    Distinct functions of histone H3, lysine 4 methyltransferases in normal and malignant hematopoiesis

    Curr. Opin. Hematol.

    (2017)
  • A.V. Krivtsov et al.

    MLL translocations, histone modifications and leukaemia stem-cell development

    Nat. Rev. Canc.

    (2007)
  • A.V. Krivtsov et al.

    Mixed-Lineage leukemia fusions and chromatin in leukemia

    Cold Spring Harb Perspect Med

    (2017)
  • Cited by (102)

    View all citing articles on Scopus
    View full text