1
|
Rørvik SD, Torkildsen S, Bruserud Ø, Tvedt THA. Acute myeloid leukemia with rare recurring translocations-an overview of the entities included in the international consensus classification. Ann Hematol 2024; 103:1103-1119. [PMID: 38443661 PMCID: PMC10940453 DOI: 10.1007/s00277-024-05680-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 02/19/2024] [Indexed: 03/07/2024]
Abstract
Two different systems exist for subclassification of acute myeloid leukemia (AML); the World Health Organization (WHO) Classification and the International Consensus Classification (ICC) of myeloid malignancies. The two systems differ in their classification of AML defined by recurrent chromosomal abnormalities. One difference is that the ICC classification defines an AML subset that includes 12 different genetic abnormalities that occur in less than 4% of AML patients. These subtypes exhibit distinct clinical traits and are associated with treatment outcomes, but detailed description of these entities is not easily available and is not described in detail even in the ICC. We searched in the PubMed database to identify scientific publications describing AML patients with the recurrent chromosomal abnormalities/translocations included in this ICC defined patient subset. This patient subset includes AML with t(1;3)(p36.3;q21.3), t(3;5)(q25.3;q35.1), t(8;16)(p11.2;p13.3), t(1;22)(p13.3;q13.1), t(5;11)(q35.2;p15.4), t(11;12)(p15.4;p13.3) (involving NUP98), translocation involving NUP98 and other partner, t(7;12)(q36.3;p13.2), t(10;11)(p12.3;q14.2), t(16;21)(p11.2;q22.2), inv(16)(p13.3q24.3) and t(16;21)(q24.3;q22.1). In this updated review we describe the available information with regard to frequency, biological functions of the involved genes and the fusion proteins, morphology/immunophenotype, required diagnostic procedures, clinical characteristics (including age distribution) and prognostic impact for each of these 12 genetic abnormalities.
Collapse
Affiliation(s)
- Synne D Rørvik
- Department of Cardiology, Haukeland University Hospital, Bergen, Norway
| | - Synne Torkildsen
- Department of Haematology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Øystein Bruserud
- Acute Leukemia Research Group, Department of Clinical Science, University of Bergen, Bergen, Norway
- Section for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
2
|
Korbecki J, Kupnicka P, Barczak K, Bosiacki M, Ziętek P, Chlubek D, Baranowska-Bosiacka I. The Role of CXCR1, CXCR2, CXCR3, CXCR5, and CXCR6 Ligands in Molecular Cancer Processes and Clinical Aspects of Acute Myeloid Leukemia (AML). Cancers (Basel) 2023; 15:4555. [PMID: 37760523 PMCID: PMC10526350 DOI: 10.3390/cancers15184555] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Acute myeloid leukemia (AML) is a type of leukemia known for its unfavorable prognoses, prompting research efforts to discover new therapeutic targets. One area of investigation involves examining extracellular factors, particularly CXC chemokines. While CXCL12 (SDF-1) and its receptor CXCR4 have been extensively studied, research on other CXC chemokine axes in AML is less developed. This study aims to bridge that gap by providing an overview of the significance of CXC chemokines other than CXCL12 (CXCR1, CXCR2, CXCR3, CXCR5, and CXCR6 ligands and CXCL14 and CXCL17) in AML's oncogenic processes. We explore the roles of all CXC chemokines other than CXCL12, in particular CXCL1 (Gro-α), CXCL8 (IL-8), CXCL10 (IP-10), and CXCL11 (I-TAC) in AML tumor processes, including their impact on AML cell proliferation, bone marrow angiogenesis, interaction with non-leukemic cells like MSCs and osteoblasts, and their clinical relevance. We delve into how they influence prognosis, association with extramedullary AML, induction of chemoresistance, effects on bone marrow microvessel density, and their connection to French-American-British (FAB) classification and FLT3 gene mutations.
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046 Zielona Góra, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Paweł Ziętek
- Department of Orthopaedics, Traumatology and Orthopaedic Oncology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (J.K.); (P.K.); (M.B.); (D.C.)
| |
Collapse
|
3
|
Viita T, Côté J. The MOZ-BRPF1 acetyltransferase complex in epigenetic crosstalk linked to gene regulation, development, and human diseases. Front Cell Dev Biol 2023; 10:1115903. [PMID: 36712963 PMCID: PMC9873972 DOI: 10.3389/fcell.2022.1115903] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/29/2022] [Indexed: 01/12/2023] Open
Abstract
Acetylation of lysine residues on histone tails is an important post-translational modification (PTM) that regulates chromatin dynamics to allow gene transcription as well as DNA replication and repair. Histone acetyltransferases (HATs) are often found in large multi-subunit complexes and can also modify specific lysine residues in non-histone substrates. Interestingly, the presence of various histone PTM recognizing domains (reader domains) in these complexes ensures their specific localization, enabling the epigenetic crosstalk and context-specific activity. In this review, we will cover the biochemical and functional properties of the MOZ-BRPF1 acetyltransferase complex, underlining its role in normal biological processes as well as in disease progression. We will discuss how epigenetic reader domains within the MOZ-BRPF1 complex affect its chromatin localization and the histone acetyltransferase specificity of the complex. We will also summarize how MOZ-BRPF1 is linked to development via controlling cell stemness and how mutations or changes in expression levels of MOZ/BRPF1 can lead to developmental disorders or cancer. As a last touch, we will review the latest drug candidates for these two proteins and discuss the therapeutic possibilities.
Collapse
Affiliation(s)
| | - Jacques Côté
- St-Patrick Research Group in Basic Oncology, Oncology Division of Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Laval University Cancer Research Center, Quebec City, QC, Canada
| |
Collapse
|
4
|
Kayser S, Hills RK, Langova R, Kramer M, Guijarro F, Sustkova Z, Estey EH, Shaw CM, Ráčil Z, Mayer J, Zak P, Baer MR, Brunner AM, Szotkowski T, Cetkovsky P, Grimwade D, Walter RB, Burnett AK, Ho AD, Ehninger G, Müller-Tidow C, Platzbecker U, Thiede C, Röllig C, Schulz A, Warsow G, Brors B, Esteve J, Russell NH, Schlenk RF, Levis MJ. Characteristics and outcome of patients with acute myeloid leukaemia and t(8;16)(p11;p13): results from an International Collaborative Study. Br J Haematol 2021; 192:832-842. [PMID: 33529373 DOI: 10.1111/bjh.17336] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023]
Abstract
In acute myeloid leukaemia (AML) t(8;16)(p11;p13)/MYST3-CREBBP is a very rare abnormality. Previous small series suggested poor outcome. We report on 59 patients with t(8;16) within an international, collaborative study. Median age was 52 (range: 16-75) years. AML was de novo in 58%, therapy-related (t-AML) in 37% and secondary after myelodysplastic syndrome (s-AML) in 5%. Cytogenetics revealed a complex karyotype in 43%. Besides MYST3-CREBBP, whole-genome sequencing on a subset of 10 patients revealed recurrent mutations in ASXL1, BRD3, FLT3, MLH1, POLG, TP53, SAMD4B (n = 3, each), EYS, KRTAP9-1 SPTBN5 (n = 4, each), RUNX1 and TET2 (n = 2, each). Complete remission after intensive chemotherapy was achieved in 84%. Median follow-up was 5·48 years; five-year survival rate was 17%. Patients with s-/t-AML (P = 0·01) and those with complex karyotype (P = 0·04) had an inferior prognosis. Allogeneic haematopoietic cell transplantation (allo-HCT) was performed in 21 (36%) patients, including 15 in first complete remission (CR1). Allo-HCT in CR1 significantly improved survival (P = 0·04); multivariable analysis revealed that allo-HCT in CR1 was effective in de novo AML but not in patients with s-AML/t-AML and less in patients exhibiting a complex karyotype. In summary, outcomes of patients with t(8;16) are dismal with chemotherapy, and may be substantially improved with allo-HCT performed in CR1.
Collapse
Affiliation(s)
- Sabine Kayser
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany.,NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Ralitsa Langova
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Bioscience, University of Heidelberg, Heidelberg, Germany
| | - Michael Kramer
- Department of Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | | | - Zuzana Sustkova
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Elihu H Estey
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Department of Medicine, University of Washington, Seattle, WA, USA
| | - Carole M Shaw
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Department of Medicine, University of Washington, Seattle, WA, USA
| | - Zdeněk Ráčil
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic.,Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Masaryk University and University Hospital Brno, Brno, Czech Republic
| | - Pavel Zak
- 4th Department of Internal Medicine-Hematology, Faculty of Medicine, Charles University and University Hospital Hradec Králové, Hradec Králové, Czech Republic
| | - Maria R Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.,Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Tomas Szotkowski
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital Olomouc, Olomouc, Czech Republic
| | - Petr Cetkovsky
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - David Grimwade
- Department of Medical & Molecular Genetics, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Roland B Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.,Division of Hematology/Department of Medicine, University of Washington, Seattle, WA, USA.,Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Alan K Burnett
- Department of Haematology, School of Medicine, Cardiff University, Cardiff, UK
| | - Anthony D Ho
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Gerhard Ehninger
- Department of Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Christian Thiede
- Department of Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Christoph Röllig
- Department of Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Angela Schulz
- Genomics and Proteomics Core Facility High Throughput Sequencing, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gregor Warsow
- Omics IT and Data Management, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benedikt Brors
- Division Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany.,German Cancer Consortium (DKTK), Core Center Heidelberg, Heidelberg, Germany
| | | | - Nigel H Russell
- Department of Haematology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Richard F Schlenk
- NCT Trial Center, National Center of Tumor Diseases, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, Heidelberg, Germany
| | - Mark J Levis
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
5
|
Dupont L, Du L, Poulter M, Choi S, McIntosh M, Reeves MB. Src family kinase activity drives cytomegalovirus reactivation by recruiting MOZ histone acetyltransferase activity to the viral promoter. J Biol Chem 2019; 294:12901-12910. [PMID: 31273084 PMCID: PMC6721939 DOI: 10.1074/jbc.ra119.009667] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/29/2019] [Indexed: 12/25/2022] Open
Abstract
Human cytomegalovirus (HCMV) latency and reactivation rely on a complex interplay between cellular differentiation, cell signaling pathways, and viral gene functions. HCMV reactivation in dendritic cells (DCs) is triggered by IL-6 and extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase signaling. However, activation of the same pathway fails to reactivate HCMV in other myeloid cell types, despite this signaling axis being active in those cells. We hypothesized that IL-6-induced ERK activation initiates the changes in chromatin structure required for viral reactivation but that a concomitant signal is necessary to complete the changes in chromatin structure required for gene expression to occur. Using a differential phosphoproteomics approach in cells that do or do not support IL-6-induced viral reactivation, we identified the concomitant activation of an Src family kinase (SFK), hematopoietic cell kinase (HCK), specifically in DCs in response to IL-6. Pharmacological and genetic inhibition of HCK activity indicated that HCK is required for HCMV reactivation. Furthermore, the HCK/SFK activity was linked to recruitment of the monocytic leukemia zinc finger protein (MOZ) histone acetyltransferase to the viral promoter, which promoted histone acetylation after ERK-mediated histone phosphorylation. Importantly, pharmacological and genetic inhibition of MOZ activity prevented reactivation. These results provide an explanation for the selective activation of viral gene expression in DCs by IL-6, dependent on concomitant SFK and ERK signaling. They also reveal a previously unreported role for SFK activity in the regulation of chromatin structure at promoters in eukaryotic cells via MOZ histone acetyltransferase activity.
Collapse
Affiliation(s)
- Liane Dupont
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Lily Du
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Madeleine Poulter
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Stephanie Choi
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Megan McIntosh
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, Royal Free Hospital, University College London, Hampstead, London NW3 2PF, United Kingdom, Supported by Wellcome Trust Grant WT/204870/Z/16/Z. To whom correspondence should be addressed. Tel.:
44-203-1086783; E-mail:
| |
Collapse
|
6
|
Meng X, Zhao Y, Liu J, Wang L, Dong Z, Zhang T, Gu X, Zheng Z. Comprehensive analysis of histone modification-associated genes on differential gene expression and prognosis in gastric cancer. Exp Ther Med 2019; 18:2219-2230. [PMID: 31452712 DOI: 10.3892/etm.2019.7808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests that the epigenetic alterations caused by histone modifications have important roles in the genesis of gastric cancer (GC), particularly the well-studied acetylation and methylation modifications. In the present study, a Bioinformatics analysis of the expression of histone modification-associated genes in GC and normal tissues was performed by using datasets from Oncomine, the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). The clinical data of GC patients were downloaded from TCGA to determine the association between histone modification-associated gene expression and clinicopathological parameters or survival of GC. Finally, lysine acetyltransferase 2A (KAT2A), nuclear receptor coactivator 1 (NCOA1), SMYD family member 5 (SMYD5), protein arginine methyltransferase 1 (PRMT1) and PRDF1-RIZ (PR)/Su(var)3-9, enhancer-of-zeste and trithorax (SET) domain 16 (PRDM16) were screened; KAT2A, SMYD5 and PRMT1 were upregulated, while PRDM16 expression was downregulated in GC. Analysis of the GEO and Oncomine datasets revealed that NCOA1 was upregulated, which was contrary to the result obtained with the TCGA stomach adenocarcinoma dataset. Aberrant expression of KAT2A, NCOA1, SMYD5 and PRMT1 was more obvious in gastric intestinal-type adenocarcinoma; low NCOA1 expression was associated with better overall survival of GC patients [hazard ratio (HR)=0.690, 95% CI=0.570-0.840, P<0.001] and was an independent predictor for patients diagnosed with GC (HR=0.639, 95% CI=0.437-0.933, P=0.020). Correlation analysis and protein-protein interaction network analysis indicated a close association between ATAD2 and estrogen receptor 1 (ESR1), PRMT1, NCOA1 and KAT2A. In conclusion, differential expression of KAT2A, NCOA1, SMYD5, PRMT1 and PRDM16 was identified in GC vs. normal tissues, low NCOA1 expression was associated with poor survival of GC and ATAD2 may interact with ESR1 to regulate NCOA1 and PRMT1 in GC.
Collapse
Affiliation(s)
- Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Jingwei Liu
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lu Wang
- Department of Ultrasonography, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Zhe Dong
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Tao Zhang
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Xiaohu Gu
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| | - Zhichao Zheng
- Department of Gastric Surgery, Cancer Hospital of China Medical University/Liaoning Cancer Hospital and Institute, Shenyang, Liaoning 110042, P.R. China
| |
Collapse
|
7
|
Dhall A, Zee BM, Yan F, Blanco MA. Intersection of Epigenetic and Metabolic Regulation of Histone Modifications in Acute Myeloid Leukemia. Front Oncol 2019; 9:432. [PMID: 31192132 PMCID: PMC6540842 DOI: 10.3389/fonc.2019.00432] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/07/2019] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) is one of the most lethal blood cancers, accounting for close to a quarter of a million annual deaths worldwide. Even though genetically heterogeneous, all AMLs are characterized by two interrelated features—blocked differentiation and high proliferative capacity. Despite significant progress in our understanding of the molecular and genetic basis of AML, the treatment of AMLs with chemotherapeutic regimens has remained largely unchanged in the past 30 years. In this review, we will consider the role of two cellular processes, metabolism and epigenetics, in the development and progression of AML and highlight the studies that suggest an interconnection of therapeutic importance between the two. Large-scale whole-exome sequencing of AML patients has revealed the presence of mutations, translocations or duplications in several epigenetic effectors such as DNMT3, MLL, ASXL1, and TET2, often times co-occuring with mutations in metabolic enzymes such as IDH1 and IDH2. These mutations often result in impaired enzymatic activity which leads to an altered epigenetic landscape through dysregulation of chromatin modifications such as DNA methylation, histone acetylation and methylation. We will discuss the role of enzymes that are responsible for establishing these modifications, namely histone acetyl transferases (HAT), histone methyl transferases (HMT), demethylases (KDMs), and deacetylases (HDAC), and also highlight the merits and demerits of using inhibitors that target these enzymes. Furthermore, we will tie in the metabolic regulation of co-factors such as acetyl-CoA, SAM, and α-ketoglutarate that are utilized by these enzymes and examine the role of metabolic inhibitors as a treatment option for AML. In doing so, we hope to stimulate interest in this topic and help generate a rationale for the consideration of the combinatorial use of metabolic and epigenetic inhibitors for the treatment of AML.
Collapse
Affiliation(s)
- Abhinav Dhall
- Newborn Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Barry M Zee
- Newborn Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Fangxue Yan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - M Andres Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
8
|
Fratta E, Montico B, Rizzo A, Colizzi F, Sigalotti L, Dolcetti R. Epimutational profile of hematologic malignancies as attractive target for new epigenetic therapies. Oncotarget 2018; 7:57327-57350. [PMID: 27329599 PMCID: PMC5302993 DOI: 10.18632/oncotarget.10033] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/28/2016] [Indexed: 12/31/2022] Open
Abstract
In recent years, recurrent somatic mutations in epigenetic regulators have been identified in patients with hematological malignancies. Furthermore, chromosomal translocations in which the fusion protein partners are themselves epigenetic regulators or where epigenetic regulators are recruited/targeted by oncogenic fusion proteins have also been described. Evidence has accumulated showing that "epigenetic drugs" are likely to provide clinical benefits in several hematological malignancies, granting their approval for the treatment of myelodysplastic syndromes and cutaneous T-cell lymphomas. A large number of pre-clinical and clinical trials evaluating epigenetic drugs alone or in combination therapies are ongoing. The aim of this review is to provide a comprehensive summary of known epigenetic alterations and of the current use of epigenetic drugs for the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Elisabetta Fratta
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Barbara Montico
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Aurora Rizzo
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Francesca Colizzi
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Luca Sigalotti
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy
| | - Riccardo Dolcetti
- Cancer Bio-Immunotherapy Unit, Centro di Riferimento Oncologico, IRCCS, National Cancer Institute, Aviano, PN, Italy.,University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| |
Collapse
|
9
|
Marneth AE, Prange KHM, Al Hinai ASA, Bergevoet SM, Tesi N, Janssen-Megens EM, Kim B, Sharifi N, Yaspo ML, Kuster J, Sanders MA, Stoetman ECG, Knijnenburg J, Arentsen-Peters TCJM, Zwaan CM, Stunnenberg HG, van den Heuvel-Eibrink MM, Haferlach T, Fornerod M, Jansen JH, Valk PJM, van der Reijden BA, Martens JHA. C-terminal BRE overexpression in 11q23-rearranged and t(8;16) acute myeloid leukemia is caused by intragenic transcription initiation. Leukemia 2017; 32:828-836. [PMID: 28871137 DOI: 10.1038/leu.2017.280] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 07/16/2017] [Accepted: 08/10/2017] [Indexed: 01/05/2023]
Abstract
Overexpression of the BRE (brain and reproductive organ-expressed) gene defines a distinct pediatric and adult acute myeloid leukemia (AML) subgroup. Here we identify a promoter enriched for active chromatin marks in BRE intron 4 causing strong biallelic expression of a previously unknown C-terminal BRE transcript. This transcript starts with BRE intron 4 sequences spliced to exon 5 and downstream sequences, and if translated might code for an N terminally truncated BRE protein. Remarkably, the new BRE transcript was highly expressed in over 50% of 11q23/KMT2A (lysine methyl transferase 2A)-rearranged and t(8;16)/KAT6A-CREBBP cases, while it was virtually absent from other AML subsets and normal tissues. In gene reporter assays, the leukemia-specific fusion protein KMT2A-MLLT3 transactivated the intragenic BRE promoter. Further epigenome analyses revealed 97 additional intragenic promoter marks frequently bound by KMT2A in AML with C-terminal BRE expression. The corresponding genes may be part of a context-dependent KMT2A-MLLT3-driven oncogenic program, because they were higher expressed in this AML subtype compared with other groups. C-terminal BRE might be an important contributor to this program because in a case with relapsed AML, we observed an ins(11;2) fusing CHORDC1 to BRE at the region where intragenic transcription starts in KMT2A-rearranged and KAT6A-CREBBP AML.
Collapse
Affiliation(s)
- A E Marneth
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - K H M Prange
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - A S A Al Hinai
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S M Bergevoet
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - N Tesi
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - E M Janssen-Megens
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - B Kim
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - N Sharifi
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M L Yaspo
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - J Kuster
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M A Sanders
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - E C G Stoetman
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J Knijnenburg
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - T C J M Arentsen-Peters
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C M Zwaan
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - H G Stunnenberg
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| | - M M van den Heuvel-Eibrink
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - T Haferlach
- MLL Munich Leukemia Laboratory, Munich, Germany
| | - M Fornerod
- Pediatric Oncology/Hematology, Erasmus University Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - J H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - P J M Valk
- Department of Hematology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - B A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, The Netherlands
| | - J H A Martens
- Department of Molecular Biology, Faculty of Science, RIMLS, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
10
|
Dutta R, Tiu B, Sakamoto KM. CBP/p300 acetyltransferase activity in hematologic malignancies. Mol Genet Metab 2016; 119:37-43. [PMID: 27380996 DOI: 10.1016/j.ymgme.2016.06.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 02/08/2023]
Abstract
CREB binding protein (CBP) and p300 are critical regulators of hematopoiesis through both their transcriptional coactivator and acetyltransferase activities. Loss or mutation of CBP/p300 results in hematologic deficiencies in proliferation and differentiation as well as disruption of hematopoietic stem cell renewal and the microenvironment. Aberrant lysine acetylation mediated by CBP/p300 has recently been implicated in the genesis of multiple hematologic cancers. Understanding the effects of disrupting the acetyltransferase activity of CBP/p300 could pave the way for new therapeutic approaches to treat patients with these diseases.
Collapse
Affiliation(s)
- Ritika Dutta
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Bruce Tiu
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathleen M Sakamoto
- Division of Hematology/Oncology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Sun XJ, Man N, Tan Y, Nimer SD, Wang L. The Role of Histone Acetyltransferases in Normal and Malignant Hematopoiesis. Front Oncol 2015; 5:108. [PMID: 26075180 PMCID: PMC4443728 DOI: 10.3389/fonc.2015.00108] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 04/25/2015] [Indexed: 12/15/2022] Open
Abstract
Histone, and non-histone, protein acetylation plays an important role in a variety of cellular events, including the normal and abnormal development of blood cells, by changing the epigenetic status of chromatin and regulating non-histone protein function. Histone acetyltransferases (HATs), which are the enzymes responsible for histone and non-histone protein acetylation, contain p300/CBP, MYST, and GNAT family members. HATs are not only protein modifiers and epigenetic factors but also critical regulators of cell development and carcinogenesis. Here, we will review the function of HATs such as p300/CBP, Tip60, MOZ/MORF, and GCN5/PCAF in normal hematopoiesis and the pathogenesis of hematological malignancies. The inhibitors that have been developed to target HATs will also be reviewed here. Understanding the roles of HATs in normal/malignant hematopoiesis will provide the potential therapeutic targets for the hematological malignancies.
Collapse
Affiliation(s)
- Xiao-Jian Sun
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA ; Department of Cell Biology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Na Man
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA ; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Yurong Tan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA ; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA ; Department of Medicine, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Lan Wang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA ; Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine , Miami, FL , USA
| |
Collapse
|
12
|
Zhu L, Yang J, Zhao L, Yu X, Wang L, Wang F, Cai Y, Jin J. Expression of hMOF, but not HDAC4, is responsible for the global histone H4K16 acetylation in gastric carcinoma. Int J Oncol 2015; 46:2535-45. [PMID: 25873202 DOI: 10.3892/ijo.2015.2956] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/18/2015] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence suggests that the alteration of global histone H4K16 acetylation (H4K16ac) may be involved in several types of cancer. It is known that the global histone H4K16ac level in cells is controlled by several enzymes including histone acetyltransferases (HATs) and histone deacetylases (HDACs). We report in detail which particular enzyme is responsible for global reduction of histone H4K16ac in gastric cancer. Our study included 156 frozen tissue samples of primary diagnosed gastric cancer tissues and matched adjacent or normal tissues, and the gastric cancer cells SGC-7901 and MGC-803. The reverse transcription polymerase chain reaction (RT-PCR), western blot, transient transfection and siRNA knockdown approaches were used. Statistical analysis of the qRT-PCR data revealed that a significant reduction (>2-fold decreased) of hMOF expression in gastric cancer tissues in 81% (42/52) of patients. In patients with gastric cancer, downregulation of hMOF was connected to gastric cancer and tissues with pT2-T4 tumor status, lymph node metastasis and distant metastasis. Overall survival rates revealed a significant difference between the low- and high-hMOF expression groups. However, there was no significant difference by age, gender and cell differentiation. In SGC-7901 and MGC-803 gastric cancer cells, as expected, low expression of hMOF and decreased global histone H4K16ac were observed. Although we did not obtained a statistically significant high-level of HDAC4 in tumor tissues, increased HDAC4 in both gastric cancer cell lines was detected. Therefore, overexpression of hMOF and knockdown of HDAC4 experiments were carried out to investigate the potential coordinating role between hMOF and HDAC4 on global histone H4K16ac in gastric cancer. Overexpression of hMOF increased global H4K16ac in cells, however, no obvious increase of global H4K16ac in HDAC4 knockdown MGC-803 cells was observed. Histone acetyltransferase hMOF and global histone H4K16ac status might be involved in gastric cancer tumorigenic pathways. hMOF, but not HDAC4, is mainly responsible for global histone H4K16ac acetylation in gastric cancer cells.
Collapse
Affiliation(s)
- Lin Zhu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jiaxing Yang
- Department of Gastrointestinal Surgery, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Linhong Zhao
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Xue Yu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Lingyao Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Fei Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yong Cai
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jingji Jin
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
13
|
Perez-Campo FM, Costa G, Lie-a-Ling M, Kouskoff V, Lacaud G. The MYSTerious MOZ, a histone acetyltransferase with a key role in haematopoiesis. Immunology 2013; 139:161-165. [PMID: 23347099 PMCID: PMC3647182 DOI: 10.1111/imm.12072] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/11/2013] [Accepted: 01/11/2013] [Indexed: 12/28/2022] Open
Abstract
The MOnocytic leukaemia Zing finger (MOZ; MYST3 or KAT6A(1)) gene is frequently found translocated in acute myeloid leukaemia. MOZ encodes a large multidomain protein that contains, besides others, a histone acetyl transferase catalytic domain. Several studies have now established the critical function of MOZ in haematopoiesis. In this review we summarize the recent findings that underscore the relevance of the different biological activities of MOZ in the regulation of haematopoiesis.
Collapse
Affiliation(s)
- Flor M Perez-Campo
- Cancer Research UK Stem Cell Biology Group, Paterson Institute for Cancer Research, The University of Manchester, Manchester, UK.
| | | | | | | | | |
Collapse
|
14
|
|
15
|
Acute myeloid leukemia with translocation (8;16)(p11;p13) and MYST3-CREBBP rearrangement harbors a distinctive microRNA signature targeting RET proto-oncogene. Leukemia 2012; 27:595-603. [PMID: 23022987 DOI: 10.1038/leu.2012.278] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acute myeloid leukemia (AML) with t(8;16)(p11;p13) (t(8;16) AML) has unique clinico-biological characteristics, but its microRNA pattern is unknown. We analyzed 670 microRNAs in seven patients with t(8;16) AML and 113 with other AML subtypes. Hierarchical cluster analysis showed that all t(8;16) AML patients grouped in an independent cluster. Supervised analysis revealed a distinctive signature of 94-microRNAs, most of which were downregulated, including miR-21 and cluster miR-17-92. The mRNA expression analysis of two known transcription factors of these microRNAs (STAT3 and c-Myc, respectively) showed significant downregulation of STAT3 (P=0.04). A bioinformatic analysis showed that 29 of the downregulated microRNAs might be regulated by methylation; we treated a t(8;16) AML sample with 5-aza-2'-deoxycytidine (5-AZA-dC) and trichostatin A and found that 27 microRNAs were re-expressed after treatment. However, there was no difference in methylation status between t(8;16) and other AML subtypes, either overall or in the microRNA promoter. Cross-correlation of mRNA and microRNA expression identified RET as a potential target of several microRNAs. A Renilla-luciferase assay and flow cytometry after transfection with pre-microRNAs confirmed that RET is regulated by miR-218, miR-128, miR-27b, miR-15a and miR-195. In conclusion, t(8;16) AML harbors a specific microRNA signature that is partially epigenetically regulated and targets RET proto-oncogene.
Collapse
|
16
|
Verbrugge I, Johnstone RW, Bots M. Promises and challenges of anticancer drugs that target the epigenome. Epigenomics 2012; 3:547-65. [PMID: 22126246 DOI: 10.2217/epi.11.82] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The occurrence of epigenetic aberrations in cancer and their role in promoting tumorigenesis has led to the development of various small molecule inhibitors that target epigenetic enzymes. In preclinical settings, many epigenetic inhibitors demonstrate promising activity against a variety of both hematological and solid tumors. The therapeutic efficacy of those inhibitors that have entered the clinic however, is restricted predominantly to hematological malignancies. Here we outline the observed epigenetic aberrations in various types of cancer and the clinical responses to epigenetic drugs. We furthermore discuss strategies to improve the responsiveness of both hematological and solid malignancies to epigenetic drugs.
Collapse
Affiliation(s)
- Inge Verbrugge
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne 3002, Victoria, Australia
| | | | | |
Collapse
|
17
|
|
18
|
Lim EJ, Lu TX, Blanchard C, Rothenberg ME. Epigenetic regulation of the IL-13-induced human eotaxin-3 gene by CREB-binding protein-mediated histone 3 acetylation. J Biol Chem 2011; 286:13193-204. [PMID: 21325281 DOI: 10.1074/jbc.m110.210724] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The etiology of a variety of chronic inflammatory disorders has been attributed to the interaction of genetic and environmental factors. Herein, we identified a link between epigenetic regulation and IL-13-driven eotaxin-3 in the pathogenesis of chronic allergic inflammation. We first demonstrated that the cAMP-responsive element (CRE) site in the eotaxin-3 promoter affects IL-13-induced eotaxin-3 promoter activity. Furthermore, the CRE-binding protein-binding protein (CBP), a histone acetyltransferase, induced base-line and IL-13-induced eotaxin-3 promoter activity. Additionally, IL-13 treatment promoted global histone 3 acetylation as well as the formation of a complex containing CBP and STAT6 and the subsequent acetylation of histone 3 at the eotaxin-3 promoter. CBP gene silencing decreased IL-13-induced transcription of eotaxin-3. Conversely, inhibition of histone deacetylation increased IL-13-induced eotaxin-3 production. Clinical studies demonstrated markedly increased global acetylation of histone 3 in the inflamed tissue of patients with allergic inflammation. Collectively, these results identify an epigenetic mechanism involving CBP and chromatin remodeling in regulating IL-13-induced chemokine transcription.
Collapse
Affiliation(s)
- Eun Jin Lim
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | |
Collapse
|
19
|
Crosstalk between leukemia-associated proteins MOZ and MLL regulates HOX gene expression in human cord blood CD34+ cells. Oncogene 2010; 29:5019-31. [PMID: 20581860 DOI: 10.1038/onc.2010.254] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
MOZ and MLL, encoding a histone acetyltransferase (HAT) and a histone methyltransferase, respectively, are targets for recurrent chromosomal translocations found in acute myeloblastic or lymphoblastic leukemia. In MOZ (MOnocytic leukemia Zinc-finger protein)/CBP- or mixed lineage leukemia (MLL)-rearranged leukemias, abnormal levels of HOX transcription factors have been found to be critical for leukemogenesis. We show that MOZ and MLL cooperate to regulate these key genes in human cord blood CD34+ cells. These chromatin-modifying enzymes interact, colocalize and functionally cooperate, and both are recruited to multiple HOX promoters. We also found that WDR5, an adaptor protein essential for lysine 4 trimethylation of histone H3 (H3K4me3) by MLL, colocalizes and interacts with MOZ. We detected the binding of the HAT MOZ to H3K4me3, thus linking histone methylation to acetylation. In CD34+ cells, depletion of MLL causes release of MOZ from HOX promoters, which is correlated to defective histone activation marks, leading to repression of HOX gene expression and alteration of commitment of CD34+ cells into myeloid progenitors. Thus, our results unveil the role of the interaction between MOZ and MLL in CD34+ cells in which both proteins have a critical role in hematopoietic cell-fate decision, suggesting a new molecular mechanism by which MOZ or MLL deregulation leads to leukemogenesis.
Collapse
|
20
|
Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther 2009; 8:1409-20. [PMID: 19509247 DOI: 10.1158/1535-7163.mct-08-0860] [Citation(s) in RCA: 330] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Posttranslational modifications to histones affect chromatin structure and function resulting in altered gene expression and changes in cell behavior. Aberrant gene expression and altered epigenomic patterns are major features of cancer. Epigenetic changes including histone acetylation, histone methylation, and DNA methylation are now thought to play important roles in the onset and progression of cancer in numerous tumor types. Indeed dysregulated epigenetic modifications, especially in early neoplastic development, may be just as significant as genetic mutations in driving cancer development and growth. The reversal of aberrant epigenetic changes has therefore emerged as a potential strategy for the treatment of cancer. A number of compounds targeting enzymes that regulate histone acetylation, histone methylation, and DNA methylation have been developed as epigenetic therapies, with some demonstrating efficacy in hematological malignancies and solid tumors. This review highlights the roles of epigenetic modifications to histones and DNA in tumorigenesis and emerging epigenetic therapies being developed for the treatment of cancer.
Collapse
Affiliation(s)
- Leigh Ellis
- Peter MacCallum Cancer Center, St. Andrews Place, East Melbourne 3002, Australia
| | | | | |
Collapse
|
21
|
Katsumoto T, Yoshida N, Kitabayashi I. Roles of the histone acetyltransferase monocytic leukemia zinc finger protein in normal and malignant hematopoiesis. Cancer Sci 2008; 99:1523-7. [PMID: 18754862 PMCID: PMC11158476 DOI: 10.1111/j.1349-7006.2008.00865.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Histone-modified enzymes are involved in various cell functions, including proliferation, differentiation, cell death and carcinogenesis. The protein MOZ (monocytic leukemia zinc finger protein) is a Myst (MOZ, Ybf2 (Sas3), Sas2, Tip60)-type histone acetyltranseferase (HAT) that generates fusion genes, such as MOZ-TIF2, MOZ-CBP and MOZ-p300, in acute myeloid leukemia (AML) by chromosomal translocation. MOZ associates with AML1 (RUNX1), PU.1, and p53, and cooperatively activates target gene transcription. Gene targeting in mice has revealed that MOZ is essential for the generation and maintenance of hematopoietic stem cells (HSC) and for the appropriate development of myeloid, erythroid and B-lineage cell progenitors. In AML, MOZ fusion genes lead to repressed differentiation, hyper-proliferation, and self-renewal of myeloid progenitors through deregulation of MOZ-regulated target gene expression. It is therefore necessary to analyze the roles of MOZ and MOZ fusion genes in normal and malignant hematopoiesis to elucidate the mechanisms underlying and develop therapies for MOZ-related AML.
Collapse
Affiliation(s)
- Takuo Katsumoto
- Molecular Oncology Division, National Cancer Center Research Institute, Tokyo, Japan
| | | | | |
Collapse
|