Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Scand J Public Health ; 51(5): 673-681, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36941820

ABSTRACT

BACKGROUND: The burden of COVID-19 disease can be measured in terms of disability-adjusted life years (DALYs), which is composed of two components: the years of life lost through premature death (YLL) and the number of years lived with disability (YLD), adjusted for level of disability. This study measured DALYs due to COVID-19 in Sweden and compared it to the burden of other diseases. METHODS: The methodology used in the calculation of DALYs was based on the Global Burden of Disease guidelines. The number of patients diagnosed with mild/moderate, severe or critical COVID-19 and/or post-COVID-19 condition between March 2020 and October 2021 was extracted from national registries and used for YLD calculations. In addition, the numbers of death due to COVID-19 in different age groups were used for the YLL calculation. RESULTS: During the study period, 152,877 DALYs were lost to COVID-19 in Sweden, 99.3% of which was attributed to YLL. Loss of DALYs occurred mainly among the elderly, with 66.8% of DALYs attributed to individuals >70 years old. Compared to other diseases, the burden of COVID-19 in 2020 ranked as the eighth leading cause of DALY lost. CONCLUSIONS: Similar to other countries, the burden of COVID-19 in Sweden was concentrated mainly among the elderly, who contributed most of the DALY lost due to premature mortality. Yet, DALY loss remained lower for COVID-19 than for several other diseases. The contribution of YLD to DALYs lost was minimal. However empirical data on the occurrence and disability of post-COVID-19 condition are scarce, and YLD may therefore be underestimated.


Subject(s)
COVID-19 , Disabled Persons , Humans , Aged , Disability-Adjusted Life Years , Quality-Adjusted Life Years , Sweden/epidemiology , COVID-19/epidemiology , Cost of Illness
2.
Nat Cell Biol ; 25(2): 258-272, 2023 02.
Article in English | MEDLINE | ID: mdl-36635503

ABSTRACT

Precise control of activating H3K4me3 and repressive H3K27me3 histone modifications at bivalent promoters is essential for normal development and frequently corrupted in cancer. By coupling a cell surface readout of bivalent MHC class I gene expression with whole-genome CRISPR-Cas9 screens, we identify specific roles for MTF2-PRC2.1, PCGF1-PRC1.1 and Menin-KMT2A/B complexes in maintaining bivalency. Genetic loss or pharmacological inhibition of Menin unexpectedly phenocopies the effects of polycomb disruption, resulting in derepression of bivalent genes in both cancer cells and pluripotent stem cells. While Menin and KMT2A/B contribute to H3K4me3 at active genes, a separate Menin-independent function of KMT2A/B maintains H3K4me3 and opposes polycomb-mediated repression at bivalent genes. Release of KMT2A from active genes following Menin targeting alters the balance of polycomb and KMT2A at bivalent genes, facilitating gene activation. This functional partitioning of Menin-KMT2A/B complex components reveals therapeutic opportunities that can be leveraged through inhibition of Menin.


Subject(s)
Pluripotent Stem Cells , Transcription Factors , Polycomb-Group Proteins/genetics , Transcription Factors/genetics , Genome , Promoter Regions, Genetic
3.
iScience ; 25(1): 103679, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35036869

ABSTRACT

Abnormal activation of SETBP1 due to overexpression or missense mutations occurs frequently in various myeloid neoplasms and associates with poor prognosis. Direct activation of Hoxa9/Hoxa10/Myb transcription by SETBP1 and its missense mutants is essential for their transforming capability; however, the underlying epigenetic mechanisms remain elusive. We found that both SETBP1 and its missense mutant SETBP1(D/N) directly interact with histone methyltransferase MLL1. Using a combination of ChIP-seq and RNA-seq analysis in primary hematopoietic stem and progenitor cells, we uncovered extensive overlap in their genomic occupancy and their cooperation in activating many oncogenic transcription factor genes including Hoxa9/Hoxa10/Myb and a large group of ribosomal protein genes. Genetic ablation of Mll1 as well as treatment with an inhibitor of the MLL1 complex OICR-9429 abrogated Setbp1/Setbp1(D/N)-induced transcriptional activation and transformation. Thus, the MLL1 complex plays a critical role in Setbp1-induced transcriptional activation and transformation and represents a promising target for treating myeloid neoplasms with SETBP1 activation.

5.
Exp Hematol ; 100: 1-11, 2021 08.
Article in English | MEDLINE | ID: mdl-34298117

ABSTRACT

The clinical success of engineered, CD19-directed chimeric antigen receptor (CAR) T cells in relapsed, refractory B-cell acute lymphoblastic leukemia (B-ALL) has generated great enthusiasm for the use of CAR T cells in patients with cytogenetics that portend a poor prognosis with conventional cytotoxic therapies. One such group includes infants and children with mixed lineage leukemia (MLL1, KMT2A) rearrangements (MLL-r), who fare much worse than patients with low- or standard-risk B-ALL. Although early clinical trials using CD19 CAR T cells for MLL-r B-ALL produced complete remission in most patients, relapse with CD19-negative disease was a common mechanism of treatment failure. Whereas CD19neg relapse has been observed across a broad spectrum of B-ALL patients treated with CD19-directed therapy, patients with MLL-r have manifested the emergence of AML, often clonally related to the B-ALL, suggesting that the inherent heterogeneity or lineage plasticity of MLL-r B-ALL may predispose patients to a myeloid relapse. Understanding the factors that enable and drive myeloid relapse may be important to devise strategies to improve durability of remissions. In this review, we summarize clinical observations to date with MLL-r B-ALL and generally discuss lineage plasticity as a mechanism of escape from immunotherapy.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Immunotherapy, Adoptive , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Gene Rearrangement , Histone-Lysine N-Methyltransferase/immunology , Humans , Immunotherapy, Adoptive/methods , Myeloid-Lymphoid Leukemia Protein/immunology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Treatment Outcome , Tumor Escape
6.
Nat Commun ; 12(1): 2792, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990599

ABSTRACT

ASH1L histone methyltransferase plays a crucial role in the pathogenesis of different diseases, including acute leukemia. While ASH1L represents an attractive drug target, developing ASH1L inhibitors is challenging, as the catalytic SET domain adapts an inactive conformation with autoinhibitory loop blocking the access to the active site. Here, by applying fragment-based screening followed by medicinal chemistry and a structure-based design, we developed first-in-class small molecule inhibitors of the ASH1L SET domain. The crystal structures of ASH1L-inhibitor complexes reveal compound binding to the autoinhibitory loop region in the SET domain. When tested in MLL leukemia models, our lead compound, AS-99, blocks cell proliferation, induces apoptosis and differentiation, downregulates MLL fusion target genes, and reduces the leukemia burden in vivo. This work validates the ASH1L SET domain as a druggable target and provides a chemical probe to further study the biological functions of ASH1L as well as to develop therapeutic agents.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Binding Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Leukemia/drug therapy , Leukemia/enzymology , Animals , Antineoplastic Agents/chemistry , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drug Design , Drug Discovery , Enzyme Inhibitors/chemistry , Female , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogenes , Protein Domains , Recombinant Fusion Proteins/genetics
7.
Leukemia ; 35(5): 1405-1417, 2021 05.
Article in English | MEDLINE | ID: mdl-33542482

ABSTRACT

Translocations of Meningioma-1 (MN1) occur in a subset of acute myeloid leukemias (AML) and result in high expression of MN1, either as a full-length protein, or as a fusion protein that includes most of the N-terminus of MN1. High levels of MN1 correlate with poor prognosis. When overexpressed in murine hematopoietic progenitors, MN1 causes an aggressive AML characterized by an aberrant myeloid precursor-like gene expression program that shares features of KMT2A-rearranged (KMT2A-r) leukemia, including high levels of Hoxa and Meis1 gene expression. Compounds that target a critical KMT2A-Menin interaction have proven effective in KMT2A-r leukemia. Here, we demonstrate that Menin (Men1) is also critical for the self-renewal of MN1-driven AML through the maintenance of a distinct gene expression program. Genetic inactivation of Men1 led to a decrease in the number of functional leukemia-initiating cells. Pharmacologic inhibition of the KMT2A-Menin interaction decreased colony-forming activity, induced differentiation programs in MN1-driven murine leukemia and decreased leukemic burden in a human AML xenograft carrying an MN1-ETV6 translocation. Collectively, these results nominate Menin inhibition as a promising therapeutic strategy in MN1-driven leukemia.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Leukemic/genetics , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Mice, Knockout
8.
Stem Cell Reports ; 14(2): 285-299, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31951812

ABSTRACT

The Mixed Lineage Leukemia (MLL1, KMT2A) gene is critical for development and maintenance of hematopoietic stem cells (HSCs), however, whether this protein is limiting for HSC development is unknown due to lack of physiologic model systems. Here, we develop an MLL1-inducible embryonic stem cell (ESC) system and show that induction of wild-type MLL1 during ESC differentiation selectively increases hematopoietic potential from a transitional c-Kit+/Cd41+ population in the embryoid body and also at sites of hematopoiesis in embryos. Single-cell sequencing analysis illustrates inherent heterogeneity of the c-Kit+/Cd41+ population and demonstrates that MLL1 induction shifts its composition toward multilineage hematopoietic identities. Surprisingly, this does not occur through increasing Hox or other canonical MLL1 targets but through an enhanced Rac/Rho/integrin signaling state, which increases responsiveness to Vla4 ligands and enhances hematopoietic commitment. Together, our data implicate a Rac/Rho/integrin signaling axis in the endothelial to hematopoietic transition and demonstrate that MLL1 actives this axis.


Subject(s)
Hematopoiesis , Histone-Lysine N-Methyltransferase/metabolism , Integrins/metabolism , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Signal Transduction , rac GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Adhesion , Cell Differentiation , Colony-Forming Units Assay , Embryoid Bodies/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mesoderm/cytology , Mice
9.
Nat Commun ; 10(1): 4256, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31534153

ABSTRACT

PAX7 is a paired-homeobox transcription factor that specifies the myogenic identity of muscle stem cells and acts as a nodal factor by stimulating proliferation while inhibiting differentiation. We previously found that PAX7 recruits the H3K4 methyltransferases MLL1/2 to epigenetically activate target genes. Here we report that in the absence of Mll1, myoblasts exhibit reduced H3K4me3 at both Pax7 and Myf5 promoters and reduced Pax7 and Myf5 expression. Mll1-deficient myoblasts fail to proliferate but retain their differentiation potential, while deletion of Mll2 had no discernable effect. Re-expression of PAX7 in committed Mll1 cKO myoblasts restored H3K4me3 enrichment at the Myf5 promoter and Myf5 expression. Deletion of Mll1 in satellite cells reduced satellite cell proliferation and self-renewal, and significantly impaired skeletal muscle regeneration. Pax7 expression was unaffected in quiescent satellite cells but was markedly downregulated following satellite cell activation. Therefore, MLL1 is required for PAX7 expression and satellite cell function in vivo. Furthermore, PAX7, but not MLL1, is required for Myf5 transcriptional activation in committed myoblasts.


Subject(s)
Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myoblasts/metabolism , Myogenic Regulatory Factor 5/metabolism , PAX7 Transcription Factor/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Female , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myogenic Regulatory Factor 5/genetics , PAX7 Transcription Factor/genetics , Promoter Regions, Genetic/genetics
10.
Genes Dev ; 33(17-18): 1265-1279, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31395741

ABSTRACT

Chromosomal rearrangements of the mixed lineage leukemia (MLL) gene occur in ∼10% of B-cell acute lymphoblastic leukemia (B-ALL) and define a group of patients with dismal outcomes. Immunohistochemical staining of bone marrow biopsies from most of these patients revealed aberrant expression of BCL6, a transcription factor that promotes oncogenic B-cell transformation and drug resistance in B-ALL. Our genetic and ChIP-seq (chromatin immunoprecipitation [ChIP] combined with high-throughput sequencing) analyses showed that MLL-AF4 and MLL-ENL fusions directly bound to the BCL6 promoter and up-regulated BCL6 expression. While oncogenic MLL fusions strongly induced aberrant BCL6 expression in B-ALL cells, germline MLL was required to up-regulate Bcl6 in response to physiological stimuli during normal B-cell development. Inducible expression of Bcl6 increased MLL mRNA levels, which was reversed by genetic deletion and pharmacological inhibition of Bcl6, suggesting a positive feedback loop between MLL and BCL6. Highlighting the central role of BCL6 in MLL-rearranged B-ALL, conditional deletion and pharmacological inhibition of BCL6 compromised leukemogenesis in transplant recipient mice and restored sensitivity to vincristine chemotherapy in MLL-rearranged B-ALL patient samples. Oncogenic MLL fusions strongly induced transcriptional activation of the proapoptotic BH3-only molecule BIM, while BCL6 was required to curb MLL-induced expression of BIM. Notably, peptide (RI-BPI) and small molecule (FX1) BCL6 inhibitors derepressed BIM and synergized with the BH3-mimetic ABT-199 in eradicating MLL-rearranged B-ALL cells. These findings uncover MLL-dependent transcriptional activation of BCL6 as a previously unrecognized requirement of malignant transformation by oncogenic MLL fusions and identified BCL6 as a novel target for the treatment of MLL-rearranged B-ALL.


Subject(s)
Gene Expression Regulation, Leukemic , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Survival/genetics , Cells, Cultured , Gene Deletion , Gene Targeting , Humans , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prognosis , Promoter Regions, Genetic/genetics
11.
Cell Cycle ; 18(14): 1525-1531, 2019 07.
Article in English | MEDLINE | ID: mdl-31161857

ABSTRACT

The deregulation of hematopoietic stem cell (HSC) transcriptional networks is a common theme in acute myelogenous leukemia (AML). Chromosomal translocations that alter the Mixed Lineage Leukemia 1 gene (MLL1, MLL, KMT2A) occur in infant, childhood and adult leukemia and at the same time, wild-type MLL1 is a critical regulator of HSC homeostasis. Typically, the endogenous, wild-type (WT) MLL1 and MLL fusion oncoproteins (MLL-FPs) remain both expressed in leukemia. WT and MLL-FPs activate overlapping sets of target genes, presenting a challenge for the selective therapeutic targeting of leukemic cells. We previously demonstrated that endogenous MLL1 is not required for the maintenance of MLL-FP-driven AML but is required for normal HSC homeostasis. Here we address the role of MLL-FPs in the initiation of leukemia in the absence of endogenous MLL1. We show that loss of endogenous Mll1 results in a rapid decrease in expression of shared HSC/leukemia target genes, yet MLL-AF9 restores the expression of most of these target genes in the absence of WT MLL1, with the critical exception of Mecom/Evi1. These observations underscore the sufficiency of MLL-fusion oncoproteins for initiating leukemia, but also illustrate that WT MLL1 target genes differ in their ability to be re-activated by MLL-FPs.


Subject(s)
Hematopoietic Stem Cells/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Female , Gene Expression Regulation, Leukemic , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Humans , Leukemia, Myeloid, Acute/metabolism , MDS1 and EVI1 Complex Locus Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology
12.
Exp Hematol ; 69: 37-42, 2019 01.
Article in English | MEDLINE | ID: mdl-30315824

ABSTRACT

Disrupting the protein-protein interaction for molecularly targeted cancer therapeutics can be a challenging but promising strategy. Compounds that disrupt the interaction between menin, a chromatin-binding protein, and oncogenic mixed lineage leukemia fusion proteins (MLL-FPs) have shown significant promise in preclinical models of leukemia and have a high degree of selectivity for leukemia versus normal hematopoietic cells. Biochemical and structural studies demonstrate that, in addition to disrupting the menin-MLL-FP interaction, such compounds also inhibit menin-MLL1, menin-MLL2, and other menin-interacting proteins. Here, we address the degree to which disruption of menin-MLL-FP interactions or menin-MLL1/MLL2 interactions contribute to the antileukemia effect of menin inhibition. We show that Men1 deletion in MLL-AF9-transformed leukemia cells produces distinct cellular and molecular consequences compared with Mll1;Mll2 co-deletion and that compounds disrupting menin-MLL N-terminal interactions largely phenocopy menin loss. Moreover, we show that Mll1;Mll2-deficient leukemia cells exhibit enhanced sensitivity to menin interaction inhibitors, which is consistent with each regulating complementary genetic pathways. These data illustrate the heightened dependency of MLL-FPs on menin compared with wild-type MLL1/MLL2 for regulation of downstream target genes and argue that the predominant action of menin inhibitory compounds is through direct inhibition of MLL-FPs without significant contribution from MLL1/MLL2 inhibition.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Rearrangement , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Proto-Oncogene Proteins/genetics
13.
J Immunol ; 200(5): 1682-1691, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29351999

ABSTRACT

B lymphocyte differentiation is an exquisitely regulated homeostatic process resulting in continuous production of appropriately selected B cells. Relatively small changes in gene expression can result in deregulation of this process, leading to acute lymphocytic leukemia (ALL), immune deficiency, or autoimmunity. Translocation of MLL1 (KMT2A) often results in a pro-B cell ALL, but little is known about its role in normal B cell differentiation. Using a Rag1-cre mouse knock-in to selectively delete Mll1 in developing lymphocytes, we show that B cell, but not T cell, homeostasis depends on MLL1. Mll1-/- B progenitors fail to differentiate efficiently through the pro- to pre-B cell transition, resulting in a persistent reduction in B cell populations. Cells inefficiently transit the pre-BCR checkpoint, despite normal to higher levels of pre-BCR components, and rearranged IgH expression fails to rescue this differentiation block. Instead of IgH-rearrangement defects, we find that Mll1-/- pre-B cells exhibit attenuated RAS/MAPK signaling downstream of the pre-BCR, which results in reduced survival in physiologic levels of IL-7. Genome-wide expression data illustrate that MLL1 is connected to B cell differentiation and IL-7-dependent survival through a complex transcriptional network. Overall, our data demonstrate that wild-type MLL1 is a regulator of pre-BCR signaling and B cell differentiation and further suggest that targeting its function in pro-B cell ALL may be more broadly effective than previously anticipated.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Histone-Lysine N-Methyltransferase/metabolism , Interleukin-7/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Cells, Cultured , Female , Homeostasis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Signal Transduction/immunology
14.
Cancer Cell ; 31(6): 755-770.e6, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28609655

ABSTRACT

The MLL1 histone methyltransferase gene undergoes many distinct chromosomal rearrangements to yield poor-prognosis leukemia. The remaining wild-type allele is most commonly, but not always, retained. To what extent the wild-type allele contributes to leukemogenesis is unclear. Here we show, using rigorous, independent animal models, that endogenous MLL1 is dispensable for MLL-rearranged leukemia. Potential redundancy was addressed by co-deleting the closest paralog, Mll2. Surprisingly, Mll2 deletion alone had a significant impact on survival of MLL-AF9-transformed cells, and additional Mll1 loss further reduced viability and proliferation. We show that MLL1/MLL2 collaboration is not through redundancy, but regulation of distinct pathways. These findings highlight the relevance of MLL2 as a drug target in MLL-rearranged leukemia and suggest its broader significance in AML.


Subject(s)
DNA-Binding Proteins/genetics , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid, Acute/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Animals , Cell Proliferation/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Gene Deletion , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/physiology , Humans , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/metabolism , Neoplasm Proteins/physiology , Recombinant Fusion Proteins/genetics
15.
Curr Opin Hematol ; 24(4): 322-328, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28375985

ABSTRACT

PURPOSE OF REVIEW: Histone H3, lysine 4 (H3K4) methylation is one chromatin modification that defines distinct regulatory states of euchromatin. Mammals express six main histone methyltransferase (HMT) enzymes that modify H3K4 by monomethylation, dimethylation or trimethylation. Recent studies examine roles of some of these HMTs and their cofactors in hematopoiesis and leukemia. We discuss these emerging studies together with prior embryonic stem data, revealing how these enzymes function. RECENT FINDINGS: Murine models have been employed to conditionally or constitutively knockout HMTs (MLL1/KMT2A, MLL2/KMT2B, MLL3/KMT2C, MLL4/KMT2D, SETD1A/KMT2F and SETD1B/KMT2G) as well as specific domains or partners of these enzymes in normal hematopoietic populations and in the context of hematologic malignancies. These studies demonstrate that global or gene-specific changes in H3K4 modification levels can be attributed to particular enzymes in particular tissues. SUMMARY: Loss-of-function studies indicate largely nonoverlapping roles of the six H3K4 HMTs. These roles are not all necessarily due to differences in enzymatic activity and are not always accompanied by large global changes in histone modification. Both gain-of-function and loss-of-function mutations in hematologic malignancy are restricted to MLL1 and MLL3/MLL4, but emerging data indicate that SETD1A/SETD1B and MLL2 can be critical in leukemia as well.


Subject(s)
Hematologic Neoplasms/metabolism , Hematopoiesis , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Animals , Enhancer Elements, Genetic , Gene Expression Regulation , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Methylation , Multigene Family , Organ Specificity , Promoter Regions, Genetic , Protein Binding
16.
Int J Hematol ; 105(1): 7-16, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27796741

ABSTRACT

Accumulating recent evidence supports the notion that many enzymes that modify histones are fundamental players in normal hematopoiesis as well as hematologic malignancies, and represent an important new class of drug targets. Histone H3 lysine 4 (H3K4) methylation plays several distinct roles in gene expression and is modulated by specific methyltransferases and demethylases. Recent progress has been made clarifying the unique biological roles of the enzymes that carry out H3K4 methylation, yet a detailed understanding of H3K4 methylation states in various genomic contexts and the diverse functions of the enzymes that perform these methylation events is incomplete, but developing rapidly. In this review, we summarize and discuss the general mechanisms of H3K4 methylation, and how the six main enzymes from the SET/MLL family (responsible for H3K4me1/me2/me3) function in hematopoiesis and in hematologic malignancies.


Subject(s)
Hematopoiesis , Histone Chaperones/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins , Humans , Leukemia/pathology , Methylation , Protein Interaction Maps
17.
Cancer Biol Ther ; 17(12): 1274-1281, 2016 12.
Article in English | MEDLINE | ID: mdl-27801610

ABSTRACT

The reported incidence of pancreatic neuroendocrine tumors (PanNETs) has increased, due in large part to improvements in detection and awareness. However, therapeutic options are limited and a critical need exists for understanding a more thorough characterization of the molecular pathology underlying this disease. The Men1 knockout mouse model recapitulates the early stage of human PanNET development and can serve as a foundation for the development of advanced mouse models that are necessary for preclinical testing. Menin, the product of the MEN1 gene, has been shown to physically interact with the KMT2A and KMT2B histone methyltransferases. Both the KMT2A and MEN1 genes are located on chromosome 11q, which frequently undergoes loss of heterozygosity (LOH) in PanNETs. We report herein that inactivation of Kmt2a in Men1-deficient mice accelerated pancreatic islet tumorigenesis and shortened the average life span. Increases in cell proliferation were observed in mouse pancreatic islet tumors upon inactivation of both Kmt2a and Men1. The Kmt2a/Men1 double knockout mouse model can be used as a mouse model to study advanced PanNETs.


Subject(s)
Carcinogenesis/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Animals , Cell Proliferation , Islets of Langerhans/physiopathology , Loss of Heterozygosity , Mice , Mice, Knockout , Neoplasms, Experimental , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics
18.
Cancer Cell ; 30(6): 863-878, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27889185

ABSTRACT

The nucleoporin 98 gene (NUP98) is fused to a variety of partner genes in multiple hematopoietic malignancies. Here, we demonstrate that NUP98 fusion proteins, including NUP98-HOXA9 (NHA9), NUP98-HOXD13 (NHD13), NUP98-NSD1, NUP98-PHF23, and NUP98-TOP1 physically interact with mixed lineage leukemia 1 (MLL1) and the non-specific lethal (NSL) histone-modifying complexes. Chromatin immunoprecipitation sequencing illustrates that NHA9 and MLL1 co-localize on chromatin and are found associated with Hox gene promoter regions. Furthermore, MLL1 is required for the proliferation of NHA9 cells in vitro and in vivo. Inactivation of MLL1 leads to decreased expression of genes bound by NHA9 and MLL1 and reverses a gene expression signature found in NUP98-rearranged human leukemias. Our data reveal a molecular dependency on MLL1 function in NUP98-fusion-driven leukemogenesis.


Subject(s)
Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/genetics , Leukemia/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/metabolism , Animals , Cell Proliferation , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Nuclear Pore Complex Proteins/metabolism , Promoter Regions, Genetic , Tumor Cells, Cultured
19.
Neuropsychopharmacology ; 41(13): 3103-3113, 2016 12.
Article in English | MEDLINE | ID: mdl-27485686

ABSTRACT

Lysine (K) methyltransferase 2a (Kmt2a) and other regulators of H3 lysine 4 methylation, a histone modification enriched at promoters and enhancers, are widely expressed throughout the brain, but molecular and cellular phenotypes in subcortical areas remain poorly explored. We report that Kmt2a conditional deletion in postnatal forebrain is associated with excessive nocturnal activity and with absent or blunted responses to stimulant and dopaminergic agonist drugs, in conjunction with near-complete loss of spike-timing-dependent long-term potentiation in medium spiny neurons (MSNs). Selective ablation of Kmt2a, but not the ortholog Kmt2b, in adult ventral striatum/nucleus accumbens neurons markedly increased anxiety scores in multiple behavioral paradigms. Striatal transcriptome sequencing in adult mutants identified 262 Kmt2a-sensitive genes, mostly downregulated in Kmt2a-deficient mice. Transcriptional repression includes the 5-Htr2a serotonin receptor, strongly associated with anxiety- and depression-related disorders in human and animal models. Consistent with the role of Kmt2a in promoting gene expression, the transcriptional regulators Bahcc1, Isl1, and Sp9 were downregulated and affected by H3K4 promoter hypomethylation. Therefore, Kmt2a regulates synaptic plasticity in striatal neurons and provides an epigenetic drug target for anxiety and dopamine-mediated behaviors.


Subject(s)
Action Potentials/genetics , Anxiety , Dopamine Agents/pharmacology , Histone-Lysine N-Methyltransferase/deficiency , Myeloid-Lymphoid Leukemia Protein/deficiency , Neuronal Plasticity/genetics , Neurons/physiology , Ventral Striatum/cytology , Action Potentials/drug effects , Animals , Animals, Newborn , Anxiety/drug therapy , Anxiety/genetics , Anxiety/metabolism , Anxiety/physiopathology , Circadian Rhythm/drug effects , Circadian Rhythm/genetics , Disease Models, Animal , Female , Histone-Lysine N-Methyltransferase/genetics , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oligonucleotide Array Sequence Analysis , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Cancer Cell ; 30(1): 3-5, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27411579

ABSTRACT

In this issue of Cancer Cell, Stavropoulou et al. report that expression of the MLL-AF9 fusion results in acute myelogenous leukemia (AML) with different behaviors depending on cell context, which leads them to identify a transcriptional signature surprisingly resembling that of the epithelial-to-mesenchymal (EMT) transition, correlating with aggressiveness of disease.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Humans , Leukemia, Myeloid, Acute/genetics
SELECTION OF CITATIONS
SEARCH DETAIL