Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Apr 1.
Published in final edited form as: Expert Opin Ther Targets. 2019 Feb 26;23(4):267–280. doi: 10.1080/14728222.2019.1580692

Biology and targeting of the Jumonji-domain histone demethylase family in childhood neoplasia: a preclinical overview.

Tyler S McCann 1, Lays M Sobral 1, Chelsea Self 2, Joseph Hsieh 3, Marybeth Sechler 1,4, Paul Jedlicka 1,3,4,*
PMCID: PMC6509558  NIHMSID: NIHMS1521702  PMID: 30759030

Abstract

Introduction:

Epigenetic mechanisms of gene regulatory control play fundamental roles in developmental morphogenesis, and, as more recently appreciated, are heavily implicated in the onset and progression of neoplastic disease, including cancer. Many epigenetic mechanisms are therapeutically targetable, providing additional incentive for understanding of their contribution to cancer and other types of neoplasia.

Areas covered:

The Jumonji-domain histone demethylase (JHDM) family exemplifies many of the above traits. This review summarizes the current state of knowledge of the functions and pharmacologic targeting of JHDMs in cancer and other neoplastic processes, with an emphasis on diseases affecting the pediatric population.

Expert opinion:

To date, the JHDM family has largely been studied in the context of normal development and adult cancers. In contrast, comparatively few studies have addressed JHDM biology in cancer and other neoplastic diseases of childhood, especially solid (non-hematopoietic) neoplasms. Encouragingly, the few available examples support important roles for JHDMs in pediatric neoplasia, as well as potential roles for JHDM pharmacologic inhibition in disease management. Further investigations of JHDMs in cancer and other types of neoplasia of childhood can be expected to both enlighten disease biology and inform new approaches to improve disease outcomes.

Keywords: Jumonji, demethylase, epigenetics, cancer, neoplasia, pediatric

1. Introduction

Epigenetic mechanisms, long known to play key roles in development, have more recently emerged as playing diverse and important roles in the initiation and progression of cancer. In classical genetic terms, an epigenetic mechanism refers to a change in cellular phenotype that is not due to an alteration in DNA sequence, or genetic material. On a mechanistic level, epigenetic changes entail stable alterations in genomic output, or gene expression, due to changes in the chemical composition or/and structural conformation of chromatin. Chromatin, the structural and functional unit of the cellular genome, consists of repeating units of DNA wound around an octameric complex of histones, which together comprise the nucleosome. The DNA and protein components of chromatin each undergo dynamic chemical modifications. These include methylation and demethylation of DNA, and a variety of post-translational modifications involving specific residues in unstructured “tails” of the core histones. Such modifications collectively constitute the “histone code”, which helps control the expression states of associated genes.

DNA and histone modifications are deposited by enzymes collectively termed “writers”, and removed by enzymes termed “erasers” or “editors”. The resulting information is in turn interpreted by protein factors termed “readers”, in the form of differential interactions with variously modified forms of chromatin. An additional class of protein factors, the “remodelers”, through both post-translational histone modifications and ATP-dependent mechanisms, affects the packing and positioning of nucleosomes on chromatin. Many epigenetic modifying factors are endowed with multi-functionality by virtue of possessing multiple functional domains, such as a writer/eraser domain and reader domain(s). Together, this dynamic machinery of epigenetic modifiers plays a critical role in the gene expression output and phenotypic identity of the cell, with wide-ranging impacts on normal development and homeostasis.

This regulatory machinery is itself commonly dysregulated in cancer 13. Alterations in epigenetic control in cancer are diverse, ranging from mutation and altered expression of the various epigenetic modifiers and their cofactors, to mutations in constituent histones themselves. Changes in gene expression effected by these alterations impact all steps and processes of cancer initiation and progression. Epigenetic mechanisms of cancer initiation and progression are gradually being unraveled. Since many epigenetic factors are therapeutically targetable, such efforts have the potential to unlock new treatments for cancer.

Epigenetic mechanisms in cancer have been the subject of many recent reviews. The purpose of this review is to summarize and update the state of knowledge of the biology and targeting of a large, and potentially druggable, family of histone mark “erasers” – the Jumonji-domain histone demethylase (JHDM) family – in cancer and other types of neoplasia, with emphasis on neoplasms of childhood. Epigenetic mechanisms have emerged as playing particularly prominent roles in pediatric cancers and other neoplasms, which typically carry low mutational burdens 4, 5. Consequently, understanding of such mechanisms is likely to have very important prognostic and therapeutic implications in this age group.

The JHDM family contains roughly 20 genes/proteins in humans, which share a Jumonji-C (JmjC) demethylase domain (and thus druggable enzymatic activity) 610. The JmjC domain carries out demethylation of histones, and in some cases other proteins, using Fe2+ and α-ketoglutarate as cofactors. This mechanism is distinct from that utilized by the proteins LSD1 and LSD2, which constitute the second demethylase family in humans. JHDMs can be further divided into subfamilies with unique, as well as some overlapping, specificities for different histone methyl marks (summarized in Table 1). Reflecting a feature of many epigenetic modifiers noted above, JHDMs contain additional conserved functional domains with nucleic acid-binding, protein-protein interaction, chromatin reader or/and other motifs (also summarized in Table 1).

Table 1. JHDM structural/functional domains and histone mark specificity.

PTM: post-translational modification; DM: demethylase; Prot: protein; Chrom: chromatin; JmjC: Jumonji-C; ARID: AT-rich interaction domain; GATAL: GATA-like; ZF: zinc finger; JmjN: Jumonji-N; LRR: leucine-rich repeat; TPR: tetratricopeptide repeat; PHD: plant homeodomain.

Structural/functional domains Histone PTM specificity
DM DNA and RNA interactions Prot-Prot interactions Chrom reader Other H3K4 H3K9 H3K27 H3K36 other
Alternative names JmjC ARID GATAL ZF-C2HC4 ZF-C5HC2 ZF-CXXC F-box JmjN LRR TPR PHD Tudor PLU
KDM2 KDM2A FBXL11 JHDM1A × × × × × me1/2
KDM2B FBXL10 JHDM1B × × × × × me3 me1/2 H3K79me2/3
KDM3 KDM3A JMJD1A JHDM2A × × me1/2
KDM3B JMJD1B JHDM2B × × me1/2 H4R3me1/2
KDM3C JMJD1C JHDM2C × ×
KDM4 KDM4A JMJD2A JHDM3A × × × × me2/3 me2/3 H1.4K26me2/3
KDM4B JMJD2B JHDM3B × × × × me2/3 me2/3 H1.4K26me2/3
KDM4C JMJD2C JHDM3C × × × × me2/3 me2/3 H1.4K26me2/3
KDM4D JMJD2D JHDM3D × × me2/3 H1.4K26me2/3
KDM4E JMJD2E × × me2/3 H1.4K26me2/3
KDM5 KDM5A JARID1A RBBP2 × × × × × × me2/3
KDM5B JARID1B PLU-1 × × × × × × me2/3
KDM5C JARID1C SMCX × × × × × × me2/3
KDM5D JARID1D SMCY × × × × × × me2/3
KDM6 KDM6A UTX × × × me2/3
KDM6B JMJD3 × × me2/3
KDM6C UTY × × ×
KDM7 KDM7A KIAA1718 JHDM1D × × me1/2 me1/2
KDM7B PHF8 JHDM1F × × me1/2 me2 H4K20me1
KDM7C PHF2 JHDM1E × × me2 me2

As true of most, and likely all, epigenetic regulators, the roles of JHDMs in cancer initiation and progression show context-specificity. That said, many JHDMs have been demonstrated to exert disease-promoting roles in cancer. Based on such findings, the development of pharmacologic inhibitors of the JHDM family, both broadly acting and specific, has been an area of substantial activity 1113. Where relevant, information on inhibitors showing preclinical promise will be discussed.

2. KDM subfamilies in cancer and other neoplastic disease

2.1. KDM2

The KDM2 subfamily consists of two genes, KDM2A (JHDM1A/FBXL11) and KDM2B (JHDM1B/FBXL10). The JmjC domain of KDM2A and KDM2B catalyzes the removal of mono- and di-methyl marks at H3K36 6, 8, 9. H3K36me2 is associated with coding regions of transcribed genes 14, and also plays a role in the recruitment of DNA repair factors to sites of DNA damage 15. H3K36 methylation has been proposed to play roles both in activation and repression of gene expression 15. The JmjC domain of KDM2B is also able to remove the tri-methyl mark at H3K4 6, 8, 9, a post-translational modification strongly associated with active promoters 14. A recent study has shown that KDM2B can additionally remove di- and tri-methyl marks from H3K79, a modification associated with active transcription and involved in DNA repair 16. Utilizing their demethylase activity, members of the KDM2 family may thus activate or repress gene expression depending on context. In addition to the JmjC demethylase domain, KDM2A and KDM2B each also possess a CXXC-type zinc finger domain, capable of DNA and RNA interactions, two different types of protein-protein interaction domains (F-box and multiple leucine-rich repeat (LRR) domains), and a Plant Homeodomain (PHD) finger chromatin reader domain 6, 8, 9.

The KDM2 subfamily plays context-dependent roles in solid as well as hematopoietic neoplastic diseases. KDM2A is overexpressed in breast cancer, and promotes stem-like properties, chemoresistance, and angiogenesis by upregulating JAG1 expression 17. KDM2A also acts as a disease-promoting factor in non-small cell lung cancer (NSCLC) by repressing HDAC3 expression, and thereby increasing the expression of cell cycle-associated and invasion-related genes 18. In contrast, KDM2A exerts disease-suppressive effects in colon cancer by demethylating p65 (RelA) leading to inhibition of NF-κB activity 19. Consistent with a disease-promoting role, KDM2B inhibits both replicative and Ras oncogene-induced senescence in fibroblasts via repression of the Ink4a/Arf tumor suppressor locus 20. Further, KDM2B is a poor prognostic factor in gastric cancer, and its knockdown induces autophagy via PI3K/Akt/mTOR inhibition in gastric cancer cells 21. In contrast, however, KDM2B inhibits cell proliferation in HeLa cells, and its expression is decreased in Glioblastoma Multiforme (GBM), a highly aggressive brain neoplasm affecting both adult and pediatric patients, relative to normal brain and less aggressive CNS neoplasms 22.

KDM2B has also been examined, and found to have context-dependent roles, in acute leukemias, which include the most common malignant neoplasias of childhood. KDM2B plays a pro-leukemic role in Acute Lymphoblastic Leukemia (ALL), a disease predominantly affecting the pediatric population, where it cooperates with polycomb and trithorax complexes to control lineage commitment 23. In Acute Myeloblastic Leukemia (AML), a malignant hematopoietic neoplasm affecting both adults and children, KDM2B is required for disease initiation and maintenance, via mechanisms that include p15Ink4b silencing 24, and action of the non-canonical Polycomb PRC1.1 complex, of which it is part 25. In the context of Ras-driven myeloid transformation, however, KDM2B plays a restrictive rather than promotional role 23.

KDM2A and KDM2B both enhance somatic cell reprogramming, via a vitamin C-dependent mechanism that suppresses senescence and increases cell proliferation 26. KDM2A and KDM2B are also both positively regulated by hypoxia inducible factor (HIF) at the mRNA level 27. It is unknown at this point how such functions might impact cancer initiation or/and progression, though one may speculate that they could be disease-promoting.

2.2. KDM3

KDM3A (JMJD1A/JHDM2A), and its two homologs KDM3B (JMJD1B/JHDM2B) and JMJD1C (JMJD1C/JHDM2C), comprise the KDM3 subfamily. The JmjC domain of KDM3A and KDM3B has specificity for removal of mono- and di-methyl marks from H3K9 6, 8, 9. The H3K9me2 mark at gene regulatory elements is associated with inactive gene expression 14, 28; biology of the H3K9me1 mark is less well understood. JMJD1C has a JmjC domain, but whether it possesses intact demethylase activity is unclear 29, 30. KDM3A has been shown to homodimerize and to use a substrate channeling mechanism to remove H3K9 methyl groups 31. Interestingly, a recent study found that KDM3B also has arginine demethylase activity, directed toward H4R3me2s (symmetric H4R3me2) and its intermediate H4R3me1 32. Like H3K9me2, H4R3me2s correlates with less active gene expression 32. Thus, by virtue of removing repressive H3K9me2, and in the case of KDM3B also H4R3me2, repressive marks, KDM3A and KDM3B utilize their demethylase activity to increase gene expression. All members of the KDM3 subfamily additionally have a zinc finger domain, with potential for DNA or/and RNA interactions 33.

KDM3A is overexpressed in a variety of adult cancers, and has been shown to promote disease progression via multiple mechanisms, including cell proliferation and survival, cell motility and invasion, stem-like properties, angiogenesis and chemotherapy resistance 3440. KDM3A also functions as an estrogen receptor (ER) cofactor in breast cancer and androgen receptor (AR) cofactor in prostate cancer 4143. As both target and cofactor of hypoxia-inducible factor (HIF1α), KDM3A additionally contributes to the cancer-modulating effects of hypoxia 36, 38, 44. In hematopoietic neoplasms, KDM3A has been shown to promote cell survival in multiple myeloma via a KLF4-IRF2 axis 45. Interestingly, in contrast to the above disease-promoting roles in most cancers, KDM3A behaves as a tumor suppressor in germ cell neoplasms of the testis 46, diseases affecting both the adult and pediatric population. Interestingly, the testis is the tissue in which KDM3A is normally the most strongly expressed 47. KDM3B is overexpressed and disease-promoting in ALL, via repression of cell differentiation and activation of the LMO2 oncogene 48. Interestingly and in contrast, however, KDM3B is deleted and disease-suppressive in AML 49, 50. JMJD1C has been the most extensively studied member of the KDM3 subfamily in hematopoietic neoplasms, with multiple reports demonstrating its role as a disease-promoting factor in AML, including roles in survival and stem cell renewal 51, 52, and leukemia maintenance 53. JMJD1C has also been found to be overexpressed, and to functionally behave as a disease-promoting factor, in esophageal and colorectal cancer 54, 55. However, JMJD1C also plays a role in the DNA damage response, where its function may be disease-suppressive 56. Similar to KDM3A, a splicing variant of JMJD1C, s-JMJD1C, has been shown to act as a coactivator of the androgen receptor 57.

KDM3A has been studied in two different solid malignant neoplasms of childhood. In neuroblastoma, a malignancy of peripheral nervous system origin, KDM3A is induced by the disease driver and poor prognostic factor MYCN, and itself upregulates the expression of the long noncoding RNA MALAT1 58. This pathway increases the migratory and invasive potency of neuroblastoma cells, and treatment with dimethyloxalylglycine (DMOG), an α-ketoglutarate analog with inhibitory activity against JHDMs and other dioxygenases, results both in reduced MALAT1 expression and diminished cell migration and invasion. In the aggressive bone and soft tissue cancer Ewing Sarcoma, KDM3A expression is upregulated downstream of the driver oncofusion EWS/Fli1, in part through a microRNA mechanism, resulting in promotion of colony and tumor growth 59. Furthermore, acting directly, and through induction of expression of the Ets1 transcription factor, KDM3A upregulates expression of the cell surface protein MCAM, resulting in increased cell motility and metastasis 60. Ewing Sarcoma cells are also highly sensitive to the pan-JHDM pharmacologic inhibitor JIB-04 61.

Developmentally, KDM3A is essential for spermatogenesis 62, and plays an important role in sex determination 63. Adult KDM3A-deficient mice additionally develop metabolic defects and obesity 64, 65. JMJD1C also plays a role in spermatogenesis, being required for long-term germ cell maintenance 66. In embryonic stem cells (ESCs), KDM3A is induced by Oct4 and activates the expression of pluripotency-associated genes and ESC self-renewal 67. In keeping with the promotion of stemness-related phenotypes, the KDM3 family has been shown to augment the tumorigenic potential of colorectal cancer stem-like cells 68. Interestingly, stem-like colorectal cancer cells are also susceptible to targeting by the pan-JHDM pharmacologic inhibitor JIB-04 69.

2.3. KDM4

The KDM4 (JMJD2/JHDM3) subfamily consists of six genes (KDM4A-F). Five of these (KDM4A-E) encode functional proteins, while KDM4F appears to be a pseudogene 6, 8, 9. With regard to histone mark specificity, KDM4A-C demethylate H3K9me2/3, H3K36me2/3 and H1.4K26me3 marks, while KDM4D/E are able to demethylate H3K9me2/3 and H1.4K26me3, but not H3K36me2/3 marks 6, 8, 9. H3K9me2/3 marks at gene regulatory elements are associated with inactive gene expression, and the H3K9me3 is a canonical mark of transcriptionally silent heterochromatin 14, 28. As discussed above (KDM2 subfamily), H3K36 methylation can exert context-dependent effects on gene expression and also plays a role in DNA damage and repair 15. PTMs of H1.4, a linker histone, are less well characterized, but methylation of the K26 residue appears to be repressive 70. The demethylase activities of KDM4 family members thus have the capacity to activate, as well as potentially repress, gene expression. KDM4A-C, but not KDM4D/E, additionally contain chromatin reader PHD and Tudor domains 6, 8, 9. Adding a further level of functional and regulatory complexity, KDM4A and KDM4C, but not KDM4B, form homodimers and heterodimers via the N-terminal JmjN domain, and this interaction appears to be required for their demethylase activities 33.

Investigations of the KDM4 subfamily in cancer point to disease-promoting roles in both solid and hematopoietic neoplasms. KDM4A, KDM4B or/and KDM4C are overexpressed, and play disease-promotional roles, in common adult cancers, including those of skin, breast, prostate, colon and lung 11, 71. Important mechanisms include facilitation of hormone receptor action in breast and prostate cancer, and control of cell cycle progression, DNA replication and DNA repair 9, 71, 72. KDM4A-C have each also been specifically implicated in cancer progression, namely promotion of epithelial cancer metastasis 7376. Furthermore, KDM4B and KDM4C are hypoxia-induced genes 77, and thus likely contribute to the cancer-modulating properties of varying oxygen levels in tumor cells. In hematopoietic neoplasms, KDM4B and KDM4C are upregulated in AML 78, and KDM4C cooperates with the H3K4 histone methyltransferase (HMT) PRMT1 to promote AML leukemic transformation 79. Complementing findings from human cancer studies, data from genetically engineered mouse models (GEMMs), namely analyses of the loss of H3K9 methyltransferase function, suggest that decreased H3K9 methylation is generally supportive to oncogenesis (reviewed in 11). From a pharmacologic targeting perspective, studies have shown activity of KDM4-targeting inhibitors in both solid (breast cancer) and hematopoietic (AML) neoplasms 7880. The pan-JHDM inhibitor JIB-04 also restores sensitivity to cytarabine in SETD2-mutant leukemia, presumably via inhibition of KDM4 H3K36 demethylase activity 81.

To date, investigations of the KDM4 subfamily in pediatric solid neoplasms have focused on the nervous system. In neuroblastoma, expression of KDM4B was found to correlate with that of the high-risk disease gene MYCN, and high KDM4B expression to segregate with poor disease outcome 82. KDM4B was further shown to physically associate with MYCN, and to contribute to control of the MYCN expression signature, resulting in promotion of cell and tumor growth, and impairment of differentiation. Subsequent studies used a chemical genetic approach to identify compounds with an expression signature simulating KDM4B depletion and opposing a MYCN-high signature 83. These identified HDAC inhibitors and Ciclopirox as leading candidates, and the latter (an off-patent antifungal agent) was shown to have activity against neuroblastoma cells in vitro and in xenograft models in vivo. In medulloblastoma, a malignant neoplasm of central nervous system origin, analysis of copy number alterations implicated diminished H3K9 methylation as a mechanism of disease pathogenesis, including amplification or/and overexpression of KDM4B and KDM4C 84. KDM4B and KDM4C have also been shown to be overexpressed in osteosarcoma, especially in metastatic disease, and to promote invasive properties of this cancer 85.

Developmentally, the KDM4 subfamily plays important roles in stem/progenitor cell maintenance and differentiation. KDM4B and KDM4C have both unique and overlapping functions in embryonic stem cell self-renewal, contributing to the action of key stem cell regulatory modules, including Myc, the PRC complex and Nanog circuitry, in the maintenance of stem cell identity 67, 86. KDM4A and KDM4B also play roles in neural crest specification and osteogenic differentiation, respectively 87, 88. More recently, KDM4C has been found to function as a critical molecular scaffold in the assembly of lineage-specific enhancer complexes involved in the control of cell differentiation states 89. The above mechanisms could potentially contribute to deregulated cell differentiation inherent to developmentally related neoplasms of childhood.

2.4. KDM5

The KDM5 (JARID1) subfamily of histone demethylases consists of four members, KDM5A-D, all of which remove H3K4 di- and tri-methyl (H3K4me2/3) histone marks 6, 8, 9. As discussed above (KDM2 subfamily), H3K4me3 is strongly associated with promoters of active genes 14. Through removal of this mark, the canonical role of members of the KDM5 family is therefore that of transcriptional repressors. Indeed, KDM5A (also known as JARID1A, and Retinoblastoma (Rb) Binding Protein 2 or RBP2, associates with multiple repressor complexes 9092. However, the control of gene expression by members of the KDM5 family is complex and incompletely understood, with demonstration of both repressive and activating effects in normal cells 93, as well as cancer (see below). Particularly interesting is the observation that activation of gene expression is not necessarily demethylase activity-independent, as might be predicted 93, 94. KDM5C (JARID1C/SMCX), located on the X chromosome and originally identified as a gene that escapes X-chromosome inactivation 95, has also been identified as a modulator of enhancer activity 96, 97. In addition to the JmjC domain, KDM5A-D each also have: AT-rich interaction domain (ARID) and zinc finger protein modules capable of DNA or/and RNA interactions; a JmjN domain with potential for protein-protein interactions; chromatin reader PHD domains; and a “PLU” motif of unknown function, common to the KDM5 family 6, 8, 9, 33.

The KDM5 subfamily, especially KDM5A, has been quite extensively investigated in the context of normal cell physiology revealing complex effects on the growth and differentiation of normal cells. Notably, KDM5A has been shown to be able to both activate 93 and repress 98, 99 the expression of pro-proliferative cell cycle genes during cell differentiation. KDM5A and KDM5B (JARID1B/PLU-1) have also been demonstrated to contribute to Rb-mediated gene silencing during senescence 100. KDM5A has moreover been shown to promote reprogramming of induced pluripotent stem cells (iPSCs) 101. KDM5A-D all play roles in normal development, and KDM5C has been specifically implicated in the pathogenesis of X-linked mental retardation (XLMR) syndrome 6.

All members of the KDM5 subfamily have been investigated in cancer. KDM5A has been found to be overexpressed, and disease-promoting, in a number of different cancers, including lung 102, 103, breast 104, pancreas 105, kidney 106, and stomach 107. Cancer phenotypes augmented by KDM5A include both cell/tumor growth and motile/metastatic properties, as well as promotion of stem-like properties and the epithelial-mesenchymal transition. Mechanisms entail both increased (Cyclins D1 and E1, ITGB1, TNC) and decreased (p21, p27 and p57) gene expression. Interestingly, some studies have found the promotion of growth and motility by KDM5A to be dependent on its demethylase activity 102, while other studies have found similar phenotypes to be demethylase-independent 104, 105. Whether this reflects different mechanisms/pathways in different cancers, or the complexity of KDM5A function in the same biological processes (as seen in the control of normal growth, discussed above), remains to be determined. In further support of a cancer-promoting role, KDM5A has been shown to be required for both the onset 108 and maintenance 109 of Rb-deficient tumors in genetically engineered mice. Perhaps not surprisingly, in some contexts KDM5A can play a disease-suppressive role, as shown in breast cancer cells treated with PI3K inhibitors 110. Similar to KDM5A, KDM5B is amplified and overexpressed in breast cancer, and promotes disease progression 111. KDM5C promotes cell motility and invasion in breast and hepatocellular cancer 112, 113, and is overexpressed in prostate cancer 114, but behaves as a tumor suppressor in renal cancer 115. Studies of KDM5D (JARID1D/SMCY), a KDM5C paralog located on the Y chromosome, have thus far demonstrated loss, and functionally a metastasis-suppressive role, in prostate cancer 116118.

KDM5A and KDM5B have also been implicated in chemoresistance. KDM5A is required for induction of a “drug-tolerant persister” state in non-small cell lung cancer treated with EGFR inhibitors 119, and is a mediator of temozolomide resistance in GBM 120. KDM5B marks a slow-cycling cell population in melanoma that is required for long-term tumor growth and is selected by pharmacotherapy 121. KDM5B overexpression also contributes to chemotherapy resistance in ovarian cancer 122. KDM5A and KDM5B, along with a number of other JHDMs, are additionally upregulated in taxane/platin-resistant NSCLC cells 123. Identification of the KDM5-dependent drug-tolerant persister state motivated the development of the KDM5-specific pharmacologic inhibitor CPI-455 124. This inhibitor has been shown to have efficacy against drug-resistant NSCLC, melanoma, colon, and breast cancer cells 124, as well as KDM5B-high oral cancer 125. Other inhibitors targeting the demethylase activity of the KDM5 family have also been developed, as recently reviewed 126. Moreover, the pan-JHDM inhibitor JIB-04, which shows particular potency against the KDM5 family 127, 128, has been shown to have efficacy against taxane/platin-resistant NSCLC 123 and temozolomide-resistant GBM 129.

Studies of the KDM5 subfamily in childhood neoplasms have thus far been relatively limited. KDM5B has been investigated in neuroblastoma, where its high expression segregates with poor patient outcome 130. This same study showed that KDM5B is more highly expressed in MYCN-amplified cell lines, and its expression is further enriched in tumor spheres derived from both MYCN-amplified and non-amplified cell lines. Such KDM5Bhigh tumor spheres show enhanced stem-like properties, and are more resistant to the chemotherapeutic agents doxorubicin, etoposide and cisplatin. Functionally, studies in MYCN-amplified cells show that KDM5B promotes tumor sphere forming potency, invasiveness and cisplatin resistance. Mechanistically, KDM5B promotes the epithelial-mesenchymal transition, and expression of the Notch pathway components Jagged 1 and Notch 1 and 2. More recently, KDM5A has been shown to repress p53 expression via a translational mechanism, resulting in augmented growth of p53 wild-type cancers, including neuroblastoma 131. In hematologic malignancies, mutations of KDM5C have been identified in a subset of both de novo and relapsed pediatric acute lymphoblastic leukemia (ALL) patients 132, as well as in relapsed chemorefractory pediatric acute myeloid leukemia (AML) patients 133, but the functional consequences of these mutations have not been investigated.

2.5. KDM6

The KDM6 subfamily consists of three genes, KDM6A-C. Each member has a JmjC domain with specificity for removal of di- and tri-methyl marks on Lysine 27 of histone H3 (H3K27me2/3) 6, 8, 9. H3K27me3 is associated with inactive promoters 14, and poised enhancers 134. By removing these repressive marks, members of the KDM6 family thus utilize their JmjC domain to activate gene expression. KDM6A (UTX) is located on the X chromosome, and escapes X-inactivation to be broadly expressed 135, 136. KDM6C (UTY), a KDM6A homologue located on the Y chromosome, has been shown to have only weak demethylase activity in vitro and in vivo 137. In addition to a JmjC demethylase domain, KDM6A and KDM6C each also contain tetratricopeptide repeats involved in protein-protein interactions 138. KDM6B (JMJD3), an autosomal gene with more tissue and context-specific expression, has a JmjC demethylase domain, but no tetratricopeptide repeats or other known additional functional domains. Members of the KDM6 subfamily have both demethylase activity-dependent and independent roles in development 6, 139. KDM6A and KDM6B have also been studied in the context of iPSC reprogramming, with KDM6A shown to be promotional, and KDM6B inhibitory, of this process 139.

Investigations of the KDM6 subfamily in cancer have revealed both disease-promoting and disease-suppressive roles. In T-cell ALL, a disease predominantly affecting the pediatric population, KDM6A is mutated in male patients and functionally behaves as a tumor suppressor 136, 140. Interestingly, however, in a subtype of the same disease, TAL1-driven T-cell ALL, KDM6A has been demonstrated to act as an oncogenic cofactor essential for disease maintenance 141. KDM6A mutations have also been identified in pediatric B-cell ALL, and adult cancers of colon and kidney 139. Moreover, KDM6A is mutated in group 3 and 4 subtypes of medulloblastoma, and such mutations correlate with worse prognosis 142. In breast cancer, however, KDM6A acts as a disease-promoting factor, cooperating with the estrogen receptor and the H3K4 methyltransferase MLL4 to augment gene expression driving cell growth and metastasis 143, 144. More recently, also in breast cancer, KDM6A has been further shown to play a critical role in the resolution of promoter bivalency at genes upregulated during EMT-MET transition, a process necessary for efficient metastasis 145. KDM6A has additionally been shown to be increased in expression in AML 78.

Similar to KDM6A, there is evidence in the published literature for disease-promoting as well as disease-suppressive roles of KDM6B in cancer. In T-cell ALL, KDM6B is required for leukemia initiation and maintenance 140 and its expression is upregulated in AML 146. In liver cancer, KDM6B promotes epithelial-mesenchymal transition, and stem-like and invasive phenotypes 147, while in melanoma it enhances disease progression via effects on the tumor microenvironment 148. In contrast, KDM6B has been reported to exert disease-suppressive effects in cancers of the colon, pancreas and lung 149.

A cell-active, pharmacologic inhibitor with high selectivity for the KDM6 subfamily has been developed (GSK-J4) 150. Encouragingly, this drug has shown activity against multiple pediatric malignancies. GSK-J4 inhibited T-cell ALL in both of the above-cited studies of the KDM6 family in this disease 136, 141. More recently, GSK-J4 was shown to be effective against high-risk neuroblastoma 151. Mechanistically, GSK-J4 induced tumor cell differentiation and endoplasmic reticulum stress, resulting in cell death, and showed efficacy both in context of chemoresistance and retinoic acid resistance. Further, GSK-J4 sensitized MYCN-amplified neuroblastoma to the BCL2 inhibitor venetoclax. Whether GSK-J4 acts through inhibition of KDM6A, KDM6B or both in neuroblastoma was not clarified. GSK-J4 has also been shown to be effective against pediatric brain stem glioma 152, a highly aggressive and lethal neoplasm characterized by histone H3.3 K27 mutations (most commonly K27M). Such mutations, which are heterozygous, exert a dominant effect on wild-type H3K27, resulting in reduced levels of H3K27 methylation via sequestration of the polycomb repressive complex 2 (PRC2) 140, 153, 154. Treatment of brain stem glioma cells with GSK-J4 was able to reverse this downregulation of H3K27 methylation, at least in part via inhibition of KDM6B, and inhibited cell and tumor growth 152. GSK-J4 has also been shown to be effective in diffuse large B-cell lymphoma, as a chemotherapy-sensitizing agent 155, AML 146, breast cancer 156, taxane/platin-resistant lung cancer 123, and prostate cancer 157, including its castration-resistant subtype 158.

2.6. KDM7

The KDM7 subfamily has three members, KDM7A-C. KDM7A (JHDM1D) is also known as KIAA1718, while KDM7B (JHDM1F) and KDM7C (JHDM1E) are more commonly referred to in the literature as PHF8 and PHF2, respectively. The JmjC domain of each factor has activity against repressive H3K9me2 marks, with KDM7A and PHF8 also able to demethylate H3K9me1 6, 8, 9. KDM7A is additionally able to remove repressive H3K27me1/2 marks 6, 8, 9. PHF8 displays additional demethylase activity against repressive H3K27me2, as well as H4K20me1, a mark that can be associated with active as well as repressed genes 6, 8, 9. All three factors can thus activate gene expression via their JmjC-associated demethylase activity against H3K9 and H3K27 methyl marks. PHF8 additionally has the potential to repress gene expression, via its demethylase activity against H4K20me1 in some contexts, as well as interactions with repressor complexes 159, 160. All members of the KDM7 subfamily also possess an N-terminal PHD domain, which confers the ability to differentially bind modified histones. Notably, both PHF2 and PHF8 have been shown to interact via their PHD domains with H3K4me2/3, marks typically associated with active gene regulatory elements 161, 162, suggesting a role in the regulation of genes primed for transcription. Developmentally, PHF8 plays important roles in central nervous system and craniofacial morphogenesis, and, like KDM5C, has been implicated in the pathogenesis of X-linked mental retardation (XLMR) syndrome 6.

Members of the KDM7 subfamily have been shown to play somewhat divergent roles in cancer. PHF8, thus far the most extensively studied, has been shown to be upregulated in expression and to be disease-promoting in a number of different cancers, including breast 163, 164, prostate 165, lung 166 and esophagus 167. Demonstrated functional roles for PHF8 in these cancers include augmentation of cell and tumor growth, enhancement of motile properties and metastasis, and a role in DNA repair. Mechanistically, in prostate cancer, PHF8 is induced by c-Myc and hypoxia/HIF, and functions both in concert with and downstream of AR to promote disease progression 168, 169. PHF8 also controls rRNA synthesis 161, promotes cell cycle progression by multiple mechanisms 170172, and controls cytoskeletal dynamics 173. In adult-onset ALL, PHF8 plays an oncogenic role via a positive feedback loop with MEK/ERK signaling 174. Studies of the other members of the KDM7 family in cancer have thus far been more limited, and have uncovered both disease-promoting and disease-suppressive roles. In prostate cancer, KDM7A is upregulated in expression and plays a role in AR-dependent gene expression and cell proliferation 175. However, KDM7A expression is also induced under nutrient deprivation, and its ectopic overexpression in HeLa cells inhibits xenograft growth and angiogenesis 176. In PANC-1 pancreatic cancer cells, KDM7A positively regulates E-cadherin expression, a cell junction protein typically expressed in normal epithelial cells 177. PHF2 has been shown to be downregulated in expression in colon and gastric cancer, and to promote p53-driven gene expression under genotoxic stress and chemoresponse, thus acting as a tumor suppressor in these contexts 178. In breast cancer, PHF2 had been shown to be phosphorylated by PKA and to promote cAMP-induced mesenchymal-epithelial transition (MET) 179. In Clear Cell Renal Cell Carcinoma (CCRCC), high expression of PHF2 is associated with longer survival and smaller tumor size 180.

Investigations of the KDM7 subfamily in neoplastic diseases of childhood have thus far focused on hematopoietic cancers. In Notch-driven Tc-ALL, PHF8 is part of the Notch nuclear complex and is required for Notch-mediated gene expression 181. In Acute Promyelocytic Leukemia, PHF8 functions as a cofactor of the disease-driving RARα oncofusions, and is required for a therapeutic response of the disease to all-trans retinoic acid 182. PHF2, on the other hand, is downregulated in a subset of ALL patients with deletions of the PHF2 upstream regulator Ikaros, and low PHF2 expression correlates with cell proliferation and increased levels of markers of poor prognosis 183.

Multiple approaches have been undertaken toward development of pharmacologic inhibitors of the KDM7 subfamily, all at relatively early stages. A structure-based approach identified a compound with activity against KDM7A/PHF8, and the closely related KDM2A, which inhibited proliferation of cancer cells 184. A different KDM2/7 inhibitor was identified in a screen of analogues of the JmjC domain cofactor α-ketoglutarate 185. A cyclic, cell-penetrating peptide inhibitor of PHF8 has also recently been developed 186.

3. Conclusion

Epigenetic mechanisms have long been known to play fundamental roles in normal development. As evident more recently, they are also frequently dysregulated in neoplastic disease including cancer, where they make important contributions to disease pathogenesis. Epigenetic mechanisms present potential therapeutic targets, as they frequently rely on enzymatic activities susceptible to small molecule inhibition. The Jumonji-domain histone demethylase (JHDM) family exemplifies all of the above traits. With respect to cancer and other neoplasias, at this time, the JHDM family has largely been studied in the context of adult diseases. Here, the various members have been demonstrated to exert diverse, and frequently profound, disease-modifying effects. Not surprisingly for epigenetic regulators, most JHDM demonstrate a degree of context dependence for their biological effects in cancer. However, encouragingly, from a pharmacologic targeting standpoint, a number of JHDMs (eg: KDM3A, KDM4A/B/C, KDM5A/B and PHF8) appear to behave predominantly to exclusively as disease-promoting factors. At this time, JHDMs have been far less studied in cancers and other neoplasias of childhood. However, available examples, mainly from diseases of the hematopoietic and nervous systems, support important roles, and in some cases therapeutic potential, of JHDMs in pediatric neoplasia. Further studies of JHDMs in childhood cancers and other neoplastic disorders can be expected to both inform disease biology and point to new options for disease management.

4. Expert opinion

Cancers and other neoplastic diseases of childhood commonly have a developmental basis, arising in immature developmental cell and tissue precursors, recapitulating molecular and histologic developmental programs, or manifesting both traits. Epigenetic mechanisms have long been known to play fundamental roles in development, and, more recently, their roles in the initiation and progression of neoplastic disease have come to be appreciated. It has also become apparent that epigenetic mechanisms play particularly important roles as disease drivers and promoters of disease progression in pediatric neoplasms with a developmental basis, which, in contrast to adult neoplastic processes, tend to be characterized by a relative paucity of mutations.

Genes belonging to the JHDM family first came to be known as developmental regulators 6, 10. More recently, JHDMs have emerged as important players in neoplasia and malignancy. As evident in this review, thus far, JHDMs have largely been studied in adult cancers. In this context, they have emerged as important modulators of disease progression, with predominantly disease-promoting roles. As summarized in Table 2, where studied in cancers of childhood, JHDMs have also been shown to function predominantly as disease-promoting factors. To date, in the pediatric age group, JHDMs have predominantly been investigated in hematopoietic malignancies and tumors of the nervous system. Given the important roles of JHDMs in development, it is likely that much interesting and important information remains to be uncovered regarding their roles in neoplastic processes with a developmental basis arising in other organs and tissues, including common childhood cancers of the kidney (Wilms tumor), liver (hepatoblastoma), soft tissues (rhabdomyosarcoma) and bone (Ewing Sarcoma). Encouragingly, in the handful of available examples at this time, JHDM pharmacologic inhibition has shown activity in pediatric cancers (Table 3), underscoring the importance of this endeavor.

Table 2. JHDMs implicated in cancers and other neoplasias affecting the pediatric population.

Summary of findings from published literature, as discussed in text (references in parentheses). ALL: Acute Lymphoblastic Leukemia; Bc-ALL: B-cell ALL; Tc-ALL: T-cell ALL; AML: Acute Myeloblastic Leukemia; APML: Acute Promyelocytic Leukemia; NB: Neuroblastoma; EwS: Ewing Sarcoma; OS: Osteosarcoma; MBL: Medulloblastoma; BSG: Brain Stem Glioma; GBM: Glioblastoma Multiforme; TR: Temozolomide-resistant.

Expression/alteration Function
up down mutated promoting suppressive
KDM2 KDM2A
KDM2B GBM (ref 22) ALL (ref 23), AML (refs 24, 25) AML (KRAS-driven) (ref 23)
KDM3 KDM3A NB (ref 58), EwS (ref 59) NB (ref 58), EwS (refs 59, 60)
KDM3B ALL (ref 48) AML (refs 49, 50) ALL (ref 48) AML (refs 49, 50)
KDM3C AML (refs 51, 53) AML (refs 5153)
KDM4 KDM4A
KDM4B AML (ref 78), NB (ref 82), MBL (ref 84), OS (ref 85) NB (ref 82), OS (ref 85)
KDM4C AML (ref 78), MBL (ref 84), OS (ref 85) AML (ref 79), OS (ref 85)
KDM4D
KDM4E
KDM5 KDM5A GBM(TR) (ref 120), NB (ref 131) GBM(TR) (ref 120), NB (ref 131)
KDM5B NB (ref 130) NB (ref 130)
KDM5C ALL (ref 132), AML (ref 133)
KDM5D
KDM6 KDM6A AML (ref 78) Bc-ALL (ref 139), Tc-ALL (refs 136, 140), MB (ref 142) Tc-ALL (TAD-1+) (ref 141) Tc-ALL (refs 136, 140)
KDM6B AML (ref 146) Tc-ALL (ref 140)
KDM6C
KDM7 KDM7A
PHF8 Tc-ALL (ref 181), APML (ref 182)
PHF2 ALL (ref 183) ALL (ref 183)

Table 3. Compounds with JHDM inhibitory activity that have been evaluated in cancers and other neoplasias affecting the pediatric population.

Summary of findings from published literature, as discussed in text (references in parentheses). ALL: Acute Lymphoblastic Leukemia; Tc-ALL: T-cell ALL; AML: Acute Myeloblastic Leukemia; NB: Neuroblastoma; EwS: Ewing Sarcoma; BSG: Brain Stem Glioma; GBM: Glioblastoma Multiforme; TR: Temozolomide-resistant; * with SETD2 mutation.

Drug Selectivity Disease
JIB-04 pan-JHDM ALL/AML* (ref 81), EwS (ref 61), GBM(TR) (ref 129)
DMOG ?KDM3 NB (ref 58)
Cicloprox ?KDM4 NB (ref 83)
NSC636819 KDM4A/B AML (ref 78)
SD70 KDM4C AML (refs 78, 79)
GSK-J4 KDM6 Tc-ALL (refs 136, 141), AML (refs 78, 146), NB (ref 151), BSG (ref 152)

JHDMs are most readily targetable through their JmjC domain-dependent demethylase enzymatic activity. Whether this is the optimal approach in a given cancer depends on demonstration that such activity is critical to phenotypic effects and control of expression of key downstream genes. For many JHDMs, this information remains to be determined, both in adult and pediatric cancers. Further, all JHDMs are multi-domain proteins, as summarized in Table 1. However, at this time, the role of most of these additional domains in gene regulation, and normal and abnormal biology, including cancer, largely remains to be defined. A case in point illustrating both of the above-unanswered questions is KDM5A. Canonically a repressor of gene expression via its H3K4 demethylase activity, KDM5A has been shown to be able to activate gene expression both in normal and cancer cells, including pro-metastatic genes in the case of the latter. Mechanistic studies addressing gene activation have identified potential roles for both demethylase-dependent as well as independent mechanisms. With regard to the demethylase-independent mechanisms of gene activation, the structural/functional domains responsible have, in most cases, not been defined; moreover, some studies have suggested an unexpected role for the demethylase activity in gene activation. Lastly, on a broader level, the precise role of H3K4 methylation as a determinant of gene regulatory element activity is at present incompletely understood 28. Thus, at this point, optimal strategies for therapeutic targeting of JHDMs in cancer in many cases await further information on molecular mechanisms of action.

JHDMs are one of a number of druggable epigenetic modifier families to be studied and targeted in cancer. A short, but by no means exhaustive, list of other factors and families includes the writers EZH2, DOT1L and DNMT1, the erasers LSD1 and HDACs, and the BET chromatin reader family 1. Compounds targeting these have reached clinical testing, including some for aggressive cancers of childhood, namely EZH2 inhibitors for malignant rhabdoid tumors and BET inhibitors for NUT-rearranged midline carcinoma 1. Targetable epigenetic regulators have also emerged as potential alternative approaches to inhibition of “undruggable” transcription factor oncofusions in pediatric sarcomas, such as LSD1 in Ewing Sarcoma and BET inhibitors in fusion-positive rhabdomyosarcoma 187, 188. The biology of the JHDM family in cancer is undoubtedly intertwined with these and other chromatin factor families, but the nature and relevance of such interactions is largely undefined at this time. Understanding such intersections, both at the molecular mechanistic level and at the promising lead compound level, is likely to inform new and better therapeutic strategies.

In summary and closing, the Jumonji-domain histone demethylases (JHDMs) comprise a large family of epigenetic regulators with roles in developmental morphogenesis, increasingly demonstrated roles in adult cancer initiation and progression, and, at this time, largely uncharacterized roles in the pathogenesis of neoplasms of childhood, particularly those with developmental origins. JHDMs are all druggable via their enzymatic demethylase activities, but are also multi-domain proteins, and at this time, much remains to be learned about the different domain requirements for their mechanisms of action. Similarly, much remains to be learned about JHDM mechanistic intersections with other classes of epigenetic, as well as transcriptional, regulators of gene expression. Such knowledge can be expected to inform our understanding of the mechanisms of neoplastic initiation and progression, and help guide strategies toward development of new and better therapies.

Article highlights.

- Epigenetic mechanisms of gene regulatory control play fundamental roles in developmental morphogenesis, and, as more recently appreciated, are heavily implicated in the onset and progression of neoplastic disease, including cancer. Many epigenetic mechanisms are therapeutically targetable. The Jumonji-domain histone demethylase (JHDM) family exemplifies these traits.

- The JHDM family consists of roughly 20 genes/proteins that remove methyl groups from modified histones, using the Jumonji-C (JmjC) domain, and Fe2+ and α-ketoglutarate as cofactors. Histone mark specificity varies among the different subfamily members and helps determine effects on gene expression. All JHDMs are multi-domain proteins, but, at this time, the biological functions of additional domains are much less well understood than those of the JmjC demethylase domain.

- The biology of most JHDMs has been investigated in adult cancers. Here, different JHDMs have been found to exert context-dependent effects ranging from predominantly disease-promoting to mainly disease-suppressive. Encouragingly, from a pharmacologic targeting perspective, several JHDMs appear to behave predominantly to exclusively as disease-promoting factors.

- At this time, comparatively few studies have addressed JHDM biology in cancer and other neoplastic diseases of childhood, especially solid (non-hematopoietic) neoplasms. However, the few available examples support important roles for JHDMs in pediatric neoplasia, as well as potential roles for JHDM pharmacologic inhibition in disease management.

Funding

Support for this work was provided by the National Cancer Institute (T32-CA190216 to TSM; F31-CA203053 to MS; R01-CA183874 to PJ).

Footnotes

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

References

Papers of special note, with emphasis on cancers and other neoplasias affecting the pediatric population, have been highlighted to readers (*).

  • 1.Brien GL, Valerio DG, Armstrong SA. Exploiting the Epigenome to Control Cancer-Promoting Gene-Expression Programs. Cancer Cell 2016. April 11;29(4):464–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Feinberg AP, Koldobskiy MA, Gondor A. Epigenetic modulators, modifiers and mediators in cancer aetiology and progression. Nat Rev Genet 2016. May;17(5):284–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Plass C, Pfister SM, Lindroth AM, et al. Mutations in regulators of the epigenome and their connections to global chromatin patterns in cancer. Nat Rev Genet 2013. November;14(11):765–80. [DOI] [PubMed] [Google Scholar]
  • 4.Lawlor ER, Thiele CJ. Epigenetic changes in pediatric solid tumors: promising new targets. Clin Cancer Res 2012. May 15;18(10):2768–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lawrence MS, Stojanov P, Polak P, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013. July 11;499(7457):214–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cloos PA, Christensen J, Agger K, et al. Erasing the methyl mark: histone demethylases at the center of cellular differentiation and disease. Genes Dev 2008. May 1;22(9):1115–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dimitrova E, Turberfield AH, Klose RJ. Histone demethylases in chromatin biology and beyond. EMBO Rep 2015. December;16(12):1620–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet 2012. May;13(5):343–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Johansson C, Tumber A, Che K, et al. The roles of Jumonji-type oxygenases in human disease. Epigenomics 2014. February;6(1):89–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nottke A, Colaiacovo MP, Shi Y. Developmental roles of the histone lysine demethylases. Development 2009. March;136(6):879–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hojfeldt JW, Agger K, Helin K. Histone lysine demethylases as targets for anticancer therapy. Nature reviews Drug discovery 2013. December;12(12):917–30. [DOI] [PubMed] [Google Scholar]
  • 12.Maes T, Carceller E, Salas J, et al. Advances in the development of histone lysine demethylase inhibitors. Curr Opin Pharmacol 2015. August;23:52–60. [DOI] [PubMed] [Google Scholar]
  • 13.Thinnes CC, England KS, Kawamura A, et al. Targeting histone lysine demethylases - progress, challenges, and the future. Biochim Biophys Acta 2014. December;1839(12):1416–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Li B, Carey M, Workman JL. The role of chromatin during transcription. Cell 2007. February 23;128(4):707–19. [DOI] [PubMed] [Google Scholar]
  • 15.Wagner EJ, Carpenter PB. Understanding the language of Lys36 methylation at histone H3. Nat Rev Mol Cell Biol 2012. January 23;13(2):115–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kang JY, Kim JY, Kim KB, et al. KDM2B is a histone H3K79 demethylase and induces transcriptional repression via sirtuin-1-mediated chromatin silencing. FASEB J 2018. October;32(10):5737–50. [DOI] [PubMed] [Google Scholar]
  • 17.Chen JY, Li CF, Chu PY, et al. Lysine demethylase 2A promotes stemness and angiogenesis of breast cancer by upregulating Jagged1. Oncotarget 2016. May 10;7(19):27689–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dhar SS, Alam H, Li N, et al. Transcriptional repression of histone deacetylase 3 by the histone demethylase KDM2A is coupled to tumorigenicity of lung cancer cells. J Biol Chem 2014. March 14;289(11):7483–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lu T, Jackson MW, Wang B, et al. Regulation of NF-kappaB by NSD1/FBXL11-dependent reversible lysine methylation of p65. Proc Natl Acad Sci U S A 2010. January 5;107(1):46–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Tzatsos A, Pfau R, Kampranis SC, et al. Ndy1/KDM2B immortalizes mouse embryonic fibroblasts by repressing the Ink4a/Arf locus. Proc Natl Acad Sci U S A 2009. February 24;106(8):2641–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zhao E, Tang C, Jiang X, et al. Inhibition of cell proliferation and induction of autophagy by KDM2B/FBXL10 knockdown in gastric cancer cells. Cell Signal 2017. August;36:222–29. [DOI] [PubMed] [Google Scholar]
  • 22.Frescas D, Guardavaccaro D, Bassermann F, et al. JHDM1B/FBXL10 is a nucleolar protein that represses transcription of ribosomal RNA genes. Nature 2007. November 8;450(7167):309–13. [DOI] [PubMed] [Google Scholar]
  • 23.Andricovich J, Kai Y, Peng W, et al. Histone demethylase KDM2B regulates lineage commitment in normal and malignant hematopoiesis. J Clin Invest 2016. March 1;126(3):905–20. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Characterized the functions of KDM2B in normal and malignant hematopoiesis, including divergent roles in ALL and AML.
  • 24.He J, Nguyen AT, Zhang Y. KDM2b/JHDM1b, an H3K36me2-specific demethylase, is required for initiation and maintenance of acute myeloid leukemia. Blood 2011. April 7;117(14):3869–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.van den Boom V, Maat H, Geugien M, et al. Non-canonical PRC1.1 Targets Active Genes Independent of H3K27me3 and Is Essential for Leukemogenesis. Cell reports 2016. January 12;14(2):332–46. [DOI] [PubMed] [Google Scholar]
  • 26.Wang T, Chen K, Zeng X, et al. The histone demethylases Jhdm1a/1b enhance somatic cell reprogramming in a vitamin-C-dependent manner. Cell Stem Cell 2011. December 2;9(6):575–87. [DOI] [PubMed] [Google Scholar]
  • 27.Batie M, Druker J, D’Ignazio L, et al. KDM2 Family Members are Regulated by HIF-1 in Hypoxia. Cells 2017. March 17;6(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Atlasi Y, Stunnenberg HG. The interplay of epigenetic marks during stem cell differentiation and development. Nat Rev Genet 2017. November;18(11):643–58. [DOI] [PubMed] [Google Scholar]
  • 29.Kim SM, Kim JY, Choe NW, et al. Regulation of mouse steroidogenesis by WHISTLE and JMJD1C through histone methylation balance. Nucleic Acids Res 2010. October;38(19):6389–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Brauchle M, Yao Z, Arora R, et al. Protein complex interactor analysis and differential activity of KDM3 subfamily members towards H3K9 methylation. PLoS One 2013;8(4):e60549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Goda S, Isagawa T, Chikaoka Y, et al. Control of histone H3 lysine 9 (H3K9) methylation state via cooperative two-step demethylation by Jumonji domain containing 1A (JMJD1A) homodimer. J Biol Chem 2013. December 27;288(52):36948–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li S, Ali S, Duan X, et al. JMJD1B Demethylates H4R3me2s and H3K9me2 to Facilitate Gene Expression for Development of Hematopoietic Stem and Progenitor Cells. Cell reports 2018. April 10;23(2):389–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Levin M, Stark M, Assaraf YG. The JmjN domain as a dimerization interface and a targeted inhibitor of KDM4 demethylase activity. Oncotarget 2018. March 30;9(24):16861–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cho HS, Toyokawa G, Daigo Y, et al. The JmjC domain-containing histone demethylase KDM3A is a positive regulator of the G1/S transition in cancer cells via transcriptional regulation of the HOXA1 gene. Int J Cancer 2012. August 1;131(3):E179–89. [DOI] [PubMed] [Google Scholar]
  • 35.Fan L, Peng G, Sahgal N, et al. Regulation of c-Myc expression by the histone demethylase JMJD1A is essential for prostate cancer cell growth and survival. Oncogene 2016. May 12;35(19):2441–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Krieg AJ, Rankin EB, Chan D, et al. Regulation of the histone demethylase JMJD1A by hypoxia-inducible factor 1 alpha enhances hypoxic gene expression and tumor growth. Mol Cell Biol 2010. January;30(1):344–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nakatsuka T, Tateishi K, Kudo Y, et al. Impact of histone demethylase KDM3A-dependent AP-1 transactivity on hepatotumorigenesis induced by PI3K activation. Oncogene 2017. July 10. [DOI] [PubMed] [Google Scholar]
  • 38.Uemura M, Yamamoto H, Takemasa I, et al. Jumonji domain containing 1A is a novel prognostic marker for colorectal cancer: in vivo identification from hypoxic tumor cells. Clin Cancer Res 2010. September 15;16(18):4636–46. [DOI] [PubMed] [Google Scholar]
  • 39.Ramadoss S, Guo G, Wang CY. Lysine demethylase KDM3A regulates breast cancer cell invasion and apoptosis by targeting histone and the non-histone protein p53. Oncogene 2017. January 05;36(1):47–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ramadoss S, Sen S, Ramachandran I, et al. Lysine-specific demethylase KDM3A regulates ovarian cancer stemness and chemoresistance. Oncogene 2017. March;36(11):1537–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wade MA, Jones D, Wilson L, et al. The histone demethylase enzyme KDM3A is a key estrogen receptor regulator in breast cancer. Nucleic Acids Res 2015. January;43(1):196–207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wilson S, Fan L, Sahgal N, et al. The histone demethylase KDM3A regulates the transcriptional program of the androgen receptor in prostate cancer cells. Oncotarget 2017. May 2;8(18):30328–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yamane K, Toumazou C, Tsukada Y, et al. JHDM2A, a JmjC-containing H3K9 demethylase, facilitates transcription activation by androgen receptor. Cell 2006. May 5;125(3):483–95. [DOI] [PubMed] [Google Scholar]
  • 44.Mimura I, Nangaku M, Kanki Y, et al. Dynamic change of chromatin conformation in response to hypoxia enhances the expression of GLUT3 (SLC2A3) by cooperative interaction of hypoxia-inducible factor 1 and KDM3A. Mol Cell Biol 2012. August;32(15):3018–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ohguchi H, Hideshima T, Bhasin MK, et al. The KDM3A-KLF2-IRF4 axis maintains myeloma cell survival. Nature communications 2016. January 05;7:10258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ueda J, Ho JC, Lee KL, et al. The hypoxia-inducible epigenetic regulators Jmjd1a and G9a provide a mechanistic link between angiogenesis and tumor growth. Mol Cell Biol 2014. October 1;34(19):3702–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hoog C, Schalling M, Grunder-Brundell E, et al. Analysis of a murine male germ cell-specific transcript that encodes a putative zinc finger protein. Mol Reprod Dev 1991. November;30(3):173–81. [DOI] [PubMed] [Google Scholar]
  • 48.Kim JY, Kim KB, Eom GH, et al. KDM3B is the H3K9 demethylase involved in transcriptional activation of lmo2 in leukemia. Mol Cell Biol 2012. July;32(14):2917–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hu Z, Gomes I, Horrigan SK, et al. A novel nuclear protein, 5qNCA (LOC51780) is a candidate for the myeloid leukemia tumor suppressor gene on chromosome 5 band q31. Oncogene 2001. October 18;20(47):6946–54. [DOI] [PubMed] [Google Scholar]
  • 50.Xu X, Nagel S, Quentmeier H, et al. KDM3B shows tumor-suppressive activity and transcriptionally regulates HOXA1 through retinoic acid response elements in acute myeloid leukemia. Leuk Lymphoma 2018. January;59(1):204–13. [DOI] [PubMed] [Google Scholar]
  • 51.Chen M, Zhu N, Liu X, et al. JMJD1C is required for the survival of acute myeloid leukemia by functioning as a coactivator for key transcription factors. Genes Dev 2015. October 15;29(20):2123–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhu N, Chen M, Eng R, et al. MLL-AF9- and HOXA9-mediated acute myeloid leukemia stem cell self-renewal requires JMJD1C. J Clin Invest 2016. March 1;126(3):997–1011. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Identified KDM3C as a factor that promotes AML stem cell self-renewal, but is largely dispensable for normal HSC function.
  • 53.Sroczynska P, Cruickshank VA, Bukowski JP, et al. shRNA screening identifies JMJD1C as being required for leukemia maintenance. Blood 2014. March 20;123(12):1870–82. [DOI] [PubMed] [Google Scholar]
  • 54.Ek WE, Lagergren K, Cook M, et al. Polymorphisms in genes in the androgen pathway and risk of Barrett’s esophagus and esophageal adenocarcinoma. Int J Cancer 2016. March 1;138(5):1146–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chen C, Aihemaiti M, Zhang X, et al. Downregulation of histone demethylase JMJD1C inhibits colorectal cancer metastasis through targeting ATF2. Am J Cancer Res 2018;8(5):852–65. [PMC free article] [PubMed] [Google Scholar]
  • 56.Watanabe S, Watanabe K, Akimov V, et al. JMJD1C demethylates MDC1 to regulate the RNF8 and BRCA1-mediated chromatin response to DNA breaks. Nat Struct Mol Biol 2013. December;20(12):1425–33. [DOI] [PubMed] [Google Scholar]
  • 57.Wolf SS, Patchev VK, Obendorf M. A novel variant of the putative demethylase gene, s-JMJD1C, is a coactivator of the AR. Arch Biochem Biophys 2007. April 1;460(1):56–66. [DOI] [PubMed] [Google Scholar]
  • 58.Tee AE, Ling D, Nelson C, et al. The histone demethylase JMJD1A induces cell migration and invasion by up-regulating the expression of the long noncoding RNA MALAT1. Oncotarget 2014. April 15;5(7):1793–804. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Showed that KDM3A enhances invasive properties in neuroblastoma.
  • 59.Parrish JK, Sechler M, Winn RA, et al. The histone demethylase KDM3A is a microRNA-22-regulated tumor promoter in Ewing Sarcoma. Oncogene 2015. January 8;34(2):257–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sechler M, Parrish JK, Birks DK, et al. The histone demethylase KDM3A, and its downstream target MCAM, promote Ewing Sarcoma cell migration and metastasis. Oncogene 2017. July 20;36(29):4150–60. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Identified KDM3A as a factor promoting disease progression in Ewing Sarcoma.
  • 61.Parrish JK, McCann TS, Sechler M, et al. The Jumonji-domain histone demethylase inhibitor JIB-04 deregulates oncogenic programs and increases DNA damage in Ewing Sarcoma, resulting in impaired cell proliferation and survival, and reduced tumor growth. Oncotarget 2018. September 4;9(69):33110–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Okada Y, Scott G, Ray MK, et al. Histone demethylase JHDM2A is critical for Tnp1 and Prm1 transcription and spermatogenesis. Nature 2007. November 1;450(7166):119–23. [DOI] [PubMed] [Google Scholar]
  • 63.Kuroki S, Matoba S, Akiyoshi M, et al. Epigenetic regulation of mouse sex determination by the histone demethylase Jmjd1a. Science 2013. September 6;341(6150):1106–9. [DOI] [PubMed] [Google Scholar]
  • 64.Inagaki T, Tachibana M, Magoori K, et al. Obesity and metabolic syndrome in histone demethylase JHDM2a-deficient mice. Genes Cells 2009. August;14(8):991–1001. [DOI] [PubMed] [Google Scholar]
  • 65.Tateishi K, Okada Y, Kallin EM, et al. Role of Jhdm2a in regulating metabolic gene expression and obesity resistance. Nature 2009. April 9;458(7239):757–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kuroki S, Akiyoshi M, Tokura M, et al. JMJD1C, a JmjC domain-containing protein, is required for long-term maintenance of male germ cells in mice. Biol Reprod 2013. October;89(4):93. [DOI] [PubMed] [Google Scholar]
  • 67.Loh YH, Zhang W, Chen X, et al. Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases regulate self-renewal in embryonic stem cells. Genes Dev 2007. October 15;21(20):2545–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Li J, Yu B, Deng P, et al. KDM3 epigenetically controls tumorigenic potentials of human colorectal cancer stem cells through Wnt/beta-catenin signalling. Nature communications 2017. April 25;8:15146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kim MS, Cho HI, Yoon HJ, et al. JIB-04, A Small Molecule Histone Demethylase Inhibitor, Selectively Targets Colorectal Cancer Stem Cells by Inhibiting the Wnt/beta-Catenin Signaling Pathway. Sci Rep 2018. April 26;8(1):6611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Trojer P, Zhang J, Yonezawa M, et al. Dynamic Histone H1 Isotype 4 Methylation and Demethylation by Histone Lysine Methyltransferase G9a/KMT1C and the Jumonji Domain-containing JMJD2/KDM4 Proteins. J Biol Chem 2009. March 27;284(13):8395–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Berry WL, Janknecht R. KDM4/JMJD2 histone demethylases: epigenetic regulators in cancer cells. Cancer Res 2013. May 15;73(10):2936–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell 2012. November 30;48(4):491–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ding X, Pan H, Li J, et al. Epigenetic activation of AP1 promotes squamous cell carcinoma metastasis. Sci Signal 2013. April 30;6(273):ra28 1,–13, S0–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Luo W, Chang R, Zhong J, et al. Histone demethylase JMJD2C is a coactivator for hypoxia-inducible factor 1 that is required for breast cancer progression. Proc Natl Acad Sci U S A 2012. December 4;109(49):E3367–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wilson C, Qiu L, Hong Y, et al. The histone demethylase KDM4B regulates peritoneal seeding of ovarian cancer. Oncogene 2017. May 4;36(18):2565–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Zhao L, Li W, Zang W, et al. JMJD2B promotes epithelial-mesenchymal transition by cooperating with beta-catenin and enhances gastric cancer metastasis. Clin Cancer Res 2013. December 1;19(23):6419–29. [DOI] [PubMed] [Google Scholar]
  • 77.Pollard PJ, Loenarz C, Mole DR, et al. Regulation of Jumonji-domain-containing histone demethylases by hypoxia-inducible factor (HIF)-1alpha. Biochem J 2008. December 15;416(3):387–94. [DOI] [PubMed] [Google Scholar]
  • 78.Boila LD, Chatterjee SS, Banerjee D, et al. KDM6 and KDM4 histone lysine demethylases emerge as molecular therapeutic targets in human acute myeloid leukemia. Exp Hematol 2018. February;58:44–51 e7. [DOI] [PubMed] [Google Scholar]
  • 79.Cheung N, Fung TK, Zeisig BB, et al. Targeting Aberrant Epigenetic Networks Mediated by PRMT1 and KDM4C in Acute Myeloid Leukemia. Cancer Cell 2016. January 11;29(1):32–48. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Showed that KDM4C cooperates with the histone H3K4 methyltransferase PRMT1 to promote leukemic transformation in AML, and that a KDM4C pharmacologic inhibitor has anti-leukemic activity.
  • 80.Metzger E, Stepputtis SS, Strietz J, et al. KDM4 Inhibition Targets Breast Cancer Stem-like Cells. Cancer Res 2017. November 1;77(21):5900–12. [DOI] [PubMed] [Google Scholar]
  • 81.Mar BG, Chu SH, Kahn JD, et al. SETD2 alterations impair DNA damage recognition and lead to resistance to chemotherapy in leukemia. Blood 2017. December 14;130(24):2631–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Yang J, AlTahan AM, Hu D, et al. The role of histone demethylase KDM4B in Myc signaling in neuroblastoma. J Natl Cancer Inst 2015. June;107(6):djv080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yang J, Milasta S, Hu D, et al. Targeting Histone Demethylases in MYC-Driven Neuroblastomas with Ciclopirox. Cancer Res 2017. September 1;77(17):4626–38. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Employed a chemical genetic approach to identify the off-patent drug Ciclopirox as a novel compound with activity against JHDMs and neuroblastoma cells.
  • 84.Northcott PA, Nakahara Y, Wu X, et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat Genet 2009. April;41(4):465–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Li X, Dong S. Histone demethylase JMJD2B and JMJD2C induce fibroblast growth factor 2: mediated tumorigenesis of osteosarcoma. Med Oncol 2015. Mar;32(3):53. [DOI] [PubMed] [Google Scholar]
  • 86.Das PP, Shao Z, Beyaz S, et al. Distinct and combinatorial functions of Jmjd2b/Kdm4b and Jmjd2c/Kdm4c in mouse embryonic stem cell identity. Mol Cell 2014. January 9;53(1):32–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Strobl-Mazzulla PH, Sauka-Spengler T, Bronner-Fraser M. Histone demethylase JmjD2A regulates neural crest specification. Dev Cell 2010. September 14;19(3):460–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ye L, Fan Z, Yu B, et al. Histone demethylases KDM4B and KDM6B promotes osteogenic differentiation of human MSCs. Cell Stem Cell 2012. July 6;11(1):50–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Tomaz RA, Harman JL, Karimlou D, et al. Jmjd2c facilitates the assembly of essential enhancer-protein complexes at the onset of embryonic stem cell differentiation. Development 2017. February 15;144(4):567–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liefke R, Oswald F, Alvarado C, et al. Histone demethylase KDM5A is an integral part of the core Notch-RBP-J repressor complex. Genes Dev 2010. March 15;24(6):590–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nishibuchi G, Shibata Y, Hayakawa T, et al. Physical and functional interactions between the histone H3K4 demethylase KDM5A and the nucleosome remodeling and deacetylase (NuRD) complex. J Biol Chem 2014. October 17;289(42):28956–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Pasini D, Hansen KH, Christensen J, et al. Coordinated regulation of transcriptional repression by the RBP2 H3K4 demethylase and Polycomb-Repressive Complex 2. Genes Dev 2008. May 15;22(10):1345–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Brier AB, Loft A, Madsen JG, et al. The KDM5 family is required for activation of pro-proliferative cell cycle genes during adipocyte differentiation. Nucleic Acids Res 2016. November 29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Zamurrad S, Hatch HAM, Drelon C, et al. A Drosophila Model of Intellectual Disability Caused by Mutations in the Histone Demethylase KDM5. Cell reports 2018. February 27;22(9):2359–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Wu J, Ellison J, Salido E, et al. Isolation and characterization of XE169, a novel human gene that escapes X-inactivation. Hum Mol Genet 1994. January;3(1):153–60. [DOI] [PubMed] [Google Scholar]
  • 96.Outchkourov NS, Muino JM, Kaufmann K, et al. Balancing of histone H3K4 methylation states by the Kdm5c/SMCX histone demethylase modulates promoter and enhancer function. Cell reports 2013. April 25;3(4):1071–9. [DOI] [PubMed] [Google Scholar]
  • 97.Shen H, Xu W, Guo R, et al. Suppression of Enhancer Overactivation by a RACK7-Histone Demethylase Complex. Cell 2016. April 7;165(2):331–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Beshiri ML, Holmes KB, Richter WF, et al. Coordinated repression of cell cycle genes by KDM5A and E2F4 during differentiation. Proc Natl Acad Sci U S A 2012. November 06;109(45):18499–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lopez-Bigas N, Kisiel TA, Dewaal DC, et al. Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation. Mol Cell 2008. August 22;31(4):520–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chicas A, Kapoor A, Wang X, et al. H3K4 demethylation by Jarid1a and Jarid1b contributes to retinoblastoma-mediated gene silencing during cellular senescence. Proc Natl Acad Sci U S A 2012. June 5;109(23):8971–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Pfaff N, Liebhaber S, Mobus S, et al. Inhibition of miRNA-212/132 improves the reprogramming of fibroblasts into induced pluripotent stem cells by de-repressing important epigenetic remodelling factors. Stem Cell Res 2017. April;20:70–75. [DOI] [PubMed] [Google Scholar]
  • 102.Teng YC, Lee CF, Li YS, et al. Histone demethylase RBP2 promotes lung tumorigenesis and cancer metastasis. Cancer Res 2013. August 01;73(15):4711–21. [DOI] [PubMed] [Google Scholar]
  • 103.Wang S, Wang Y, Wu H, et al. RBP2 induces epithelial-mesenchymal transition in non-small cell lung cancer. PLoS One 2013;8(12):e84735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Cao J, Liu Z, Cheung WK, et al. Histone demethylase RBP2 is critical for breast cancer progression and metastasis. Cell reports 2014. March 13;6(5):868–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Maggi EC, Trillo-Tinoco J, Struckhoff AP, et al. Retinoblastoma-binding protein 2 (RBP2) is frequently expressed in neuroendocrine tumors and promotes the neoplastic phenotype. Oncogenesis 2016. August 22;5(8):e257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zhou D, Kannappan V, Chen X, et al. RBP2 induces stem-like cancer cells by promoting EMT and is a prognostic marker for renal cell carcinoma. Exp Mol Med 2016. June 10;48:e238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zeng J, Ge Z, Wang L, et al. The histone demethylase RBP2 Is overexpressed in gastric cancer and its inhibition triggers senescence of cancer cells. Gastroenterology 2010. March;138(3):981–92. [DOI] [PubMed] [Google Scholar]
  • 108.Lin W, Cao J, Liu J, et al. Loss of the retinoblastoma binding protein 2 (RBP2) histone demethylase suppresses tumorigenesis in mice lacking Rb1 or Men1. Proc Natl Acad Sci U S A 2011. August 16;108(33):13379–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.McBrayer SK, Olenchock BA, DiNatale GJ, et al. Autochthonous tumors driven by Rb1 loss have an ongoing requirement for the RBP2 histone demethylase. Proc Natl Acad Sci U S A 2018. April 17;115(16):E3741–E48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Spangle JM, Dreijerink KM, Groner AC, et al. PI3K/AKT Signaling Regulates H3K4 Methylation in Breast Cancer. Cell reports 2016. June 21;15(12):2692–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Yamamoto S, Wu Z, Russnes HG, et al. JARID1B is a luminal lineage-driving oncogene in breast cancer. Cancer Cell 2014. June 16;25(6):762–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wang Q, Wei J, Su P, et al. Histone demethylase JARID1C promotes breast cancer metastasis cells via down regulating BRMS1 expression. Biochem Biophys Res Commun 2015. August 21;464(2):659–66. [DOI] [PubMed] [Google Scholar]
  • 113.Ji X, Jin S, Qu X, et al. Lysine-specific demethylase 5C promotes hepatocellular carcinoma cell invasion through inhibition BMP7 expression. BMC Cancer 2015. October 26;15:801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Stein J, Majores M, Rohde M, et al. KDM5C is overexpressed in prostate cancer and is a prognostic marker for prostate-specific antigen-relapse following radical prostatectomy. Am J Pathol 2014. September;184(9):2430–7. [DOI] [PubMed] [Google Scholar]
  • 115.Niu X, Zhang T, Liao L, et al. The von Hippel-Lindau tumor suppressor protein regulates gene expression and tumor growth through histone demethylase JARID1C. Oncogene 2012. February 9;31(6):776–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Komura K, Yoshikawa Y, Shimamura T, et al. ATR inhibition controls aggressive prostate tumors deficient in Y-linked histone demethylase KDM5D. J Clin Invest 2018. July 2;128(7):2979–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Li N, Dhar SS, Chen TY, et al. JARID1D Is a Suppressor and Prognostic Marker of Prostate Cancer Invasion and Metastasis. Cancer Res 2016. February 15;76(4):831–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Li N, Li Y, Lv J, et al. ZMYND8 Reads the Dual Histone Mark H3K4me1-H3K14ac to Antagonize the Expression of Metastasis-Linked Genes. Mol Cell 2016. August 4;63(3):470–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sharma SV, Lee DY, Li B, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010. April 2;141(1):69–80. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Identified an epigenetically mediated, reversible and targetable, drug-tolerant state in cancer cells, involving KDM5A.
  • 120.Banelli B, Carra E, Barbieri F, et al. The histone demethylase KDM5A is a key factor for the resistance to temozolomide in glioblastoma. Cell Cycle 2015;14(21):3418–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Roesch A, Vultur A, Bogeski I, et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 2013. June 10;23(6):811–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Wang L, Mao Y, Du G, et al. Overexpression of JARID1B is associated with poor prognosis and chemotherapy resistance in epithelial ovarian cancer. Tumour Biol 2015. April;36(4):2465–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Dalvi MP, Wang L, Zhong R, et al. Taxane-Platin-Resistant Lung Cancers Co-develop Hypersensitivity to JumonjiC Demethylase Inhibitors. Cell reports 2017. May 23;19(8):1669–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Vinogradova M, Gehling VS, Gustafson A, et al. An inhibitor of KDM5 demethylases reduces survival of drug-tolerant cancer cells. Nat Chem Biol 2016. July;12(7):531–8. [DOI] [PubMed] [Google Scholar]
  • 125.Facompre ND, Harmeyer KM, Sahu V, et al. Targeting JARID1B’s demethylase activity blocks a subset of its functions in oral cancer. Oncotarget 2018. February 6;9(10):8985–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Taylor-Papadimitriou J, Burchell J. JARID1/KDM5 demethylases as cancer targets? Expert Opin Ther Targets 2017. January;21(1):5–7. [DOI] [PubMed] [Google Scholar]
  • 127.Horton JR, Engstrom A, Zoeller EL, et al. Characterization of a Linked Jumonji Domain of the KDM5/JARID1 Family of Histone H3 Lysine 4 Demethylases. J Biol Chem 2016. February 5;291(6):2631–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wang L, Chang J, Varghese D, et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nature communications 2013;4:2035. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Initial identification of JIB-04 as a novel pan-JHDM inhibitor, now shown to have activity against a number of different cancers.
  • 129.Banelli B, Daga A, Forlani A, et al. Small molecules targeting histone demethylase genes (KDMs) inhibit growth of temozolomide-resistant glioblastoma cells. Oncotarget 2017. May 23;8(21):34896–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Kuo YT, Liu YL, Adebayo BO, et al. JARID1B Expression Plays a Critical Role in Chemoresistance and Stem Cell-Like Phenotype of Neuroblastoma Cells. PLoS One 2015;10(5):e0125343. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Showed that KDM5B promotes stem-like properties and chemoresistance in neuroblastoma.
  • 131.Hu D, Jablonowski C, Cheng PH, et al. KDM5A Regulates a Translational Program that Controls p53 Protein Expression. iScience 2018. October 17;9:84–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Mar BG, Bullinger LB, McLean KM, et al. Mutations in epigenetic regulators including SETD2 are gained during relapse in paediatric acute lymphoblastic leukaemia. Nature communications 2014. March 24;5:3469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Zhan D, Zhang Y, Xiao P, et al. Whole exome sequencing identifies novel mutations of epigenetic regulators in chemorefractory pediatric acute myeloid leukemia. Leuk Res 2018. February;65:20–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Heinz S, Romanoski CE, Benner C, et al. The selection and function of cell type-specific enhancers. Nat Rev Mol Cell Biol 2015. March;16(3):144–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Greenfield A, Carrel L, Pennisi D, et al. The UTX gene escapes X inactivation in mice and humans. Hum Mol Genet 1998. April;7(4):737–42. [DOI] [PubMed] [Google Scholar]
  • 136.Van der Meulen J, Sanghvi V, Mavrakis K, et al. The H3K27me3 demethylase UTX is a gender-specific tumor suppressor in T-cell acute lymphoblastic leukemia. Blood 2015. January 1;125(1):13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Shpargel KB, Sengoku T, Yokoyama S, et al. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet 2012. September;8(9):e1002964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Blatch GL, Lassle M. The tetratricopeptide repeat: a structural motif mediating protein-protein interactions. Bioessays 1999. November;21(11):932–9. [DOI] [PubMed] [Google Scholar]
  • 139.Arcipowski KM, Martinez CA, Ntziachristos P. Histone demethylases in physiology and cancer: a tale of two enzymes, JMJD3 and UTX. Curr Opin Genet Dev 2016. February;36:59–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ntziachristos P, Tsirigos A, Welstead GG, et al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature 2014. October 23;514(7523):513–7. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Defined contrasting roles of KDM6A (UTX) and KDM6B (JMJD3) in T-cell ALL.
  • 141.Benyoucef A, Palii CG, Wang C, et al. UTX inhibition as selective epigenetic therapy against TAL1-driven T-cell acute lymphoblastic leukemia. Genes Dev 2016. March 1;30(5):508–21. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Demonstrated pro-leukemic role of KDM6A (UTX) in the TAL-1 subtype of T-cell ALL, and anti-leukemic activity of the KDM6 inhibitor GSK-J4 in this disease.
  • 142.Dubuc AM, Remke M, Korshunov A, et al. Aberrant patterns of H3K4 and H3K27 histone lysine methylation occur across subgroups in medulloblastoma. Acta Neuropathol 2013. March;125(3):373–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Kim JH, Sharma A, Dhar SS, et al. UTX and MLL4 coordinately regulate transcriptional programs for cell proliferation and invasiveness in breast cancer cells. Cancer Res 2014. March 15;74(6):1705–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Xie G, Liu X, Zhang Y, et al. UTX promotes hormonally responsive breast carcinogenesis through feed-forward transcription regulation with estrogen receptor. Oncogene 2017. September 28;36(39):5497–511. [DOI] [PubMed] [Google Scholar]
  • 145.Taube JH, Sphyris N, Johnson KS, et al. The H3K27me3-demethylase KDM6A is suppressed in breast cancer stem-like cells, and enables the resolution of bivalency during the mesenchymal-epithelial transition. Oncotarget 2017. September 12;8(39):65548–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Li Y, Zhang M, Sheng M, et al. Therapeutic potential of GSK-J4, a histone demethylase KDM6B/JMJD3 inhibitor, for acute myeloid leukemia. J Cancer Res Clin Oncol 2018. June;144(6):1065–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Tang B, Qi G, Tang F, et al. Aberrant JMJD3 Expression Upregulates Slug to Promote Migration, Invasion, and Stem Cell-Like Behaviors in Hepatocellular Carcinoma. Cancer Res 2016. November 15;76(22):6520–32. [DOI] [PubMed] [Google Scholar]
  • 148.Park WY, Hong BJ, Lee J, et al. H3K27 Demethylase JMJD3 Employs the NF-kappaB and BMP Signaling Pathways to Modulate the Tumor Microenvironment and Promote Melanoma Progression and Metastasis. Cancer Res 2016. January 1;76(1):161–70. [DOI] [PubMed] [Google Scholar]
  • 149.Perrigue PM, Najbauer J, Barciszewski J. Histone demethylase JMJD3 at the intersection of cellular senescence and cancer. Biochim Biophys Acta 2016. April;1865(2):237–44. [DOI] [PubMed] [Google Scholar]
  • 150.Heinemann B, Nielsen JM, Hudlebusch HR, et al. Inhibition of demethylases by GSK-J1/J4. Nature 2014. October 2;514(7520):E1–2. [DOI] [PubMed] [Google Scholar]
  • 151.Lochmann TL, Powell KM, Ham J, et al. Targeted inhibition of histone H3K27 demethylation is effective in high-risk neuroblastoma. Sci Transl Med 2018. May 16;10(441). [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Showed efficacy of KDM6 inhibitor GSK-J4 against high-risk neuroblastoma.
  • 152.Hashizume R, Andor N, Ihara Y, et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat Med 2014. December;20(12):1394–6. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Showed efficacy of KDM6 inhibitor GSK-J4 against H3K27-mutant pediatric brain stem glioma.
  • 153.Bender S, Tang Y, Lindroth AM, et al. Reduced H3K27me3 and DNA hypomethylation are major drivers of gene expression in K27M mutant pediatric high-grade gliomas. Cancer Cell 2013. November 11;24(5):660–72. [DOI] [PubMed] [Google Scholar]
  • 154.Lewis PW, Muller MM, Koletsky MS, et al. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 2013. May 17;340(6134):857–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Mathur R, Sehgal L, Havranek O, et al. Inhibition of demethylase KDM6B sensitizes diffuse large B-cell lymphoma to chemotherapeutic drugs. Haematologica 2017. February;102(2):373–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yan N, Xu L, Wu X, et al. GSKJ4, an H3K27me3 demethylase inhibitor, effectively suppresses the breast cancer stem cells. Exp Cell Res 2017. October 15;359(2):405–14. [DOI] [PubMed] [Google Scholar]
  • 157.Daures M, Idrissou M, Judes G, et al. A new metabolic gene signature in prostate cancer regulated by JMJD3 and EZH2. Oncotarget 2018. May 4;9(34):23413–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Morozov VM, Li Y, Clowers MM, et al. Inhibitor of H3K27 demethylase JMJD3/UTX GSK-J4 is a potential therapeutic option for castration resistant prostate cancer. Oncotarget 2017. September 22;8(37):62131–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Asensio-Juan E, Fueyo R, Pappa S, et al. The histone demethylase PHF8 is a molecular safeguard of the IFNgamma response. Nucleic Acids Res 2017. April 20;45(7):3800–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wang J, Lin X, Wang S, et al. PHF8 and REST/NRSF co-occupy gene promoters to regulate proximal gene expression. Sci Rep 2014. May 23;4:5008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Feng W, Yonezawa M, Ye J, et al. PHF8 activates transcription of rRNA genes through H3K4me3 binding and H3K9me1/2 demethylation. Nat Struct Mol Biol 2010. April;17(4):445–50. [DOI] [PubMed] [Google Scholar]
  • 162.Wen H, Li J, Song T, et al. Recognition of histone H3K4 trimethylation by the plant homeodomain of PHF2 modulates histone demethylation. J Biol Chem 2010. March 26;285(13):9322–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Shao P, Liu Q, Maina PK, et al. Histone demethylase PHF8 promotes epithelial to mesenchymal transition and breast tumorigenesis. Nucleic Acids Res 2017. February 28;45(4):1687–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wang Q, Ma S, Song N, et al. Stabilization of histone demethylase PHF8 by USP7 promotes breast carcinogenesis. J Clin Invest 2016. June 01;126(6):2205–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Bjorkman M, Ostling P, Harma V, et al. Systematic knockdown of epigenetic enzymes identifies a novel histone demethylase PHF8 overexpressed in prostate cancer with an impact on cell proliferation, migration and invasion. Oncogene 2012. July 19;31(29):3444–56. [DOI] [PubMed] [Google Scholar]
  • 166.Shen Y, Pan X, Zhao H. The histone demethylase PHF8 is an oncogenic protein in human non-small cell lung cancer. Biochem Biophys Res Commun 2014. August 15;451(1):119–25. [DOI] [PubMed] [Google Scholar]
  • 167.Sun X, Qiu JJ, Zhu S, et al. Oncogenic features of PHF8 histone demethylase in esophageal squamous cell carcinoma. PLoS One 2013;8(10):e77353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Maina PK, Shao P, Liu Q, et al. c-MYC drives histone demethylase PHF8 during neuroendocrine differentiation and in castration-resistant prostate cancer. Oncotarget 2016. November 15;7(46):75585–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Tong D, Liu Q, Liu G, et al. The HIF/PHF8/AR axis promotes prostate cancer progression. Oncogenesis 2016. December 19;5(12):e283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Liu W, Tanasa B, Tyurina OV, et al. PHF8 mediates histone H4 lysine 20 demethylation events involved in cell cycle progression. Nature 2010. July 22;466(7305):508–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Sun L, Huang Y, Wei Q, et al. Cyclin E-CDK2 protein phosphorylates plant homeodomain finger protein 8 (PHF8) and regulates its function in the cell cycle. J Biol Chem 2015. February 13;290(7):4075–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Lim HJ, Dimova NV, Tan MK, et al. The G2/M regulator histone demethylase PHF8 is targeted for degradation by the anaphase-promoting complex containing CDC20. Mol Cell Biol 2013. November;33(21):4166–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Asensio-Juan E, Gallego C, Martinez-Balbas MA. The histone demethylase PHF8 is essential for cytoskeleton dynamics. Nucleic Acids Res 2012. October;40(19):9429–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Fu Y, Yang Y, Wang X, et al. The histone demethylase PHF8 promotes adult acute lymphoblastic leukemia through interaction with the MEK/ERK signaling pathway. Biochem Biophys Res Commun 2018. February 12;496(3):981–87. [DOI] [PubMed] [Google Scholar]
  • 175.Lee KH, Hong S, Kang M, et al. Histone demethylase KDM7A controls androgen receptor activity and tumor growth in prostate cancer. Int J Cancer 2018. September 5. [DOI] [PubMed] [Google Scholar]
  • 176.Osawa T, Muramatsu M, Wang F, et al. Increased expression of histone demethylase JHDM1D under nutrient starvation suppresses tumor growth via down-regulating angiogenesis. Proc Natl Acad Sci U S A 2011. December 20;108(51):20725–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Pan MR, Hsu MC, Chen LT, et al. G9a orchestrates PCL3 and KDM7A to promote histone H3K27 methylation. Sci Rep 2015. December 21;5:18709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Lee KH, Park JW, Sung HS, et al. PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer. Oncogene 2015. May 28;34(22):2897–909. [DOI] [PubMed] [Google Scholar]
  • 179.Pattabiraman DR, Bierie B, Kober KI, et al. Activation of PKA leads to mesenchymal-to-epithelial transition and loss of tumor-initiating ability. Science 2016. March 04;351(6277):aad3680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Lee C, Kim B, Song B, et al. Implication of PHF2 Expression in Clear Cell Renal Cell Carcinoma. J Pathol Transl Med 2017. July;51(4):359–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Yatim A, Benne C, Sobhian B, et al. NOTCH1 nuclear interactome reveals key regulators of its transcriptional activity and oncogenic function. Mol Cell 2012. November 9;48(3):445–58. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Identified PHF8 as a critical component of the Notch nuclear interactome in Notch-driven T-cell ALL.
  • 182.Arteaga MF, Mikesch JH, Qiu J, et al. The histone demethylase PHF8 governs retinoic acid response in acute promyelocytic leukemia. Cancer Cell 2013. March 18;23(3):376–89. [DOI] [PMC free article] [PubMed] [Google Scholar]; (*) Identified PHF8 as a critical cofactor of the disease-driving RARα oncofusions in APML, and showed that it is required for a therapeutic response to all-trans retinoic acid.
  • 183.Ge Z, Gu Y, Han Q, et al. Plant homeodomain finger protein 2 as a novel IKAROS target in acute lymphoblastic leukemia. Epigenomics 2018. January;10(1):59–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Suzuki T, Ozasa H, Itoh Y, et al. Identification of the KDM2/7 histone lysine demethylase subfamily inhibitor and its antiproliferative activity. J Med Chem 2013. September 26;56(18):7222–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Rose NR, Woon EC, Tumber A, et al. Plant growth regulator daminozide is a selective inhibitor of human KDM2/7 histone demethylases. J Med Chem 2012. July 26;55(14):6639–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Dorosz J, Olsen L, Seger ST, et al. Structure-Based Design of a New Scaffold for Cell-Penetrating Peptidic Inhibitors of the Histone Demethylase PHF8. Chembiochem 2017. July 18;18(14):1369–75. [DOI] [PubMed] [Google Scholar]
  • 187.Gryder BE, Yohe ME, Chou HC, et al. PAX3-FOXO1 Establishes Myogenic Super Enhancers and Confers BET Bromodomain Vulnerability. Cancer Discov 2017. August;7(8):884–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Theisen ER, Pishas KI, Saund RS, et al. Therapeutic opportunities in Ewing sarcoma: EWS-FLI inhibition via LSD1 targeting. Oncotarget 2016. April 5;7(14):17616–30. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES