Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Publication year range
1.
Blood ; 140(8): 875-888, 2022 08 25.
Article in English | MEDLINE | ID: mdl-35709354

ABSTRACT

Detailed genomic and epigenomic analyses of MECOM (the MDS1 and EVI1 complex locus) have revealed that inversion or translocation of chromosome 3 drives inv(3)/t(3;3) myeloid leukemias via structural rearrangement of an enhancer that upregulates transcription of EVI1. Here, we identify a novel, previously unannotated oncogenic RNA-splicing derived isoform of EVI1 that is frequently present in inv(3)/t(3;3) acute myeloid leukemia (AML) and directly contributes to leukemic transformation. This EVI1 isoform is generated by oncogenic mutations in the core RNA splicing factor SF3B1, which is mutated in >30% of inv(3)/t(3;3) myeloid neoplasm patients and thereby represents the single most commonly cooccurring genomic alteration in inv(3)/t(3;3) patients. SF3B1 mutations are statistically uniquely enriched in inv(3)/t(3;3) myeloid neoplasm patients and patient-derived cell lines compared with other forms of AML and promote mis-splicing of EVI1 generating an in-frame insertion of 6 amino acids at the 3' end of the second zinc finger domain of EVI1. Expression of this EVI1 splice variant enhanced the self-renewal of hematopoietic stem cells, and introduction of mutant SF3B1 in mice bearing the humanized inv(3)(q21q26) allele resulted in generation of this novel EVI1 isoform in mice and hastened leukemogenesis in vivo. The mutant SF3B1 spliceosome depends upon an exonic splicing enhancer within EVI1 exon 13 to promote usage of a cryptic branch point and aberrant 3' splice site within intron 12 resulting in the generation of this isoform. These data provide a mechanistic basis for the frequent cooccurrence of SF3B1 mutations as well as new insights into the pathogenesis of myeloid leukemias harboring inv(3)/t(3;3).


Subject(s)
Leukemia, Myeloid, Acute , Proto-Oncogenes , Animals , Chromosome Inversion , Chromosomes, Human, Pair 3/metabolism , DNA-Binding Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , MDS1 and EVI1 Complex Locus Protein/genetics , Mice , Proto-Oncogenes/genetics , Transcription Factors/metabolism
2.
Blood ; 136(14): 1670-1684, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32492700

ABSTRACT

Additional sex combs-like 1 (ASXL1), an epigenetic modulator, is frequently mutated in myeloid neoplasms. Recent analyses of mutant ASXL1 conditional knockin (ASXL1-MT-KI) mice suggested that ASXL1-MT alone is insufficient for myeloid transformation. In our previous study, we used retrovirus-mediated insertional mutagenesis, which exhibited the susceptibility of ASXL1-MT-KI hematopoietic cells to transform into myeloid leukemia cells. In this screening, we identified the hematopoietically expressed homeobox (HHEX) gene as one of the common retrovirus integration sites. In this study, we investigated the potential cooperation between ASXL1-MT and HHEX in myeloid leukemogenesis. Expression of HHEX enhanced proliferation of ASXL1-MT-expressing HSPCs by inhibiting apoptosis and blocking differentiation, whereas it showed only modest effect in normal HSPCs. Moreover, ASXL1-MT and HHEX accelerated the development of RUNX1-ETO9a and FLT3-ITD leukemia. Conversely, HHEX depletion profoundly attenuated the colony-forming activity and leukemogenicity of ASXL1-MT-expressing leukemia cells. Mechanistically, we identified MYB and ETV5 as downstream targets for ASXL1-MT and HHEX by using transcriptome and chromatin immunoprecipitation-next-generation sequencing analyses. Moreover, we found that expression of ASXL1-MT enhanced the binding of HHEX to the promoter loci of MYB or ETV5 via reducing H2AK119ub. Depletion of MYB or ETV5 induced apoptosis or differentiation in ASXL1-MT-expressing leukemia cells, respectively. In addition, ectopic expression of MYB or ETV5 reversed the reduced colony-forming activity of HHEX-depleted ASXL1-MT-expressing leukemia cells. These findings indicate that the HHEX-MYB/ETV5 axis promotes myeloid transformation in ASXL1-mutated preleukemia cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Mutation , Myeloid Cells/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Apoptosis/genetics , Biomarkers, Tumor , Biopsy , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Colony-Forming Units Assay , Disease Models, Animal , Gene Expression Profiling , Genetic Association Studies , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Immunophenotyping , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Mice , Myeloid Cells/pathology , Prognosis , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism
3.
Cancer Sci ; 112(10): 3935-3944, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34251718

ABSTRACT

Homeostasis of the hematopoietic system is achieved in a hierarchy, with hematopoietic stem cells at the pinnacle. Because only hematopoietic stem cells (HSCs) can self-renew, the size of the hematopoietic system is strictly controlled. In hematopoietic reconstitution experiments, 1 HSC can reconstitute the entire hematopoietic system, whereas 50 multipotent progenitors cannot. This indicates that only HSCs self-renew, whereas non-HSC hematopoietic progenitors are programmed to differentiate or senesce. Oncogenic mutations of the mixed lineage leukemia gene (MLL) overcome this "programmed differentiation" by conferring the self-renewing ability to non-HSC hematopoietic progenitors. In leukemia, mutated MLL proteins constitutively activate a broad range of previously transcribed CpG-rich promoters by an MLL-mediated transcriptional activation system. This system promotes self-renewal by replicating an expression profile similar to that of the mother cell in its daughter cells. In this transcriptional activation system, MLL binds to unmethylated CpG-rich promoters and recruits RNA polymerase II. MLL recruits p300/CBP through its transcriptional activation domain, which acetylates histone H3 at lysines 9, 18, and 27. The AF4 family/ENL family/P-TEFb complex (AEP) binds to acetylated H3K9/18/27 to activate transcription. Gene rearrangements of MLL with AEP- or CBP/p300-complex components generate constitutively active transcriptional machinery of this transcriptional activation system, which causes aberrant self-renewal of leukemia stem cells. Inhibitors of the components of this system effectively decrease their leukemogenic potential.


Subject(s)
Cell Self Renewal/physiology , Hematopoietic Stem Cells/physiology , Histone-Lysine N-Methyltransferase/genetics , Leukemia/etiology , Myeloid-Lymphoid Leukemia Protein/genetics , Transcriptional Activation/physiology , Acetylation , Cell Differentiation , Cell Self Renewal/genetics , Cellular Senescence , CpG Islands/genetics , DNA-Binding Proteins/metabolism , E1A-Associated p300 Protein/metabolism , Gene Rearrangement , Hematopoiesis/physiology , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Leukemia/prevention & control , Lysine/metabolism , Multipotent Stem Cells/physiology , Mutation , Myeloid-Lymphoid Leukemia Protein/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/metabolism , Transcriptional Elongation Factors/metabolism
4.
Rinsho Ketsueki ; 62(8): 988-997, 2021.
Article in Japanese | MEDLINE | ID: mdl-34497239

ABSTRACT

Leukemia is caused by uncontrolled proliferation of immature hematopoietic progenitors. MLL fusion proteins, generated by chromosomal translocations, activate a broad range of previously transcribed genes to achieve the same expression profile as that of the parent cell in the daughter cells, thereby promoting self-renewal. Normally, replication of the expression profile only occurs in the hematopoietic stem cells (HSCs). A transactivation system comprised of MLL and AF4/ENL/P-TEFb (AEP) complexes promotes it by reactivating CpG-rich promoters. In the normal hematopoietic development, this system is tightly regulated and progressively suppressed during the course of hematopoietic differentiation so that non-HSC hematopoietic cells would not self-renew. Genetic mutations such as fusions of MLL and AEP components generate a constitutively active form of the MLL transcriptional machinery, which aberrantly promotes self-renewal even in non-HSC hematopoietic cells. In this review, I depict a molecular mechanism of MLL fusion-mediated leukemogenesis from a standpoint that leukemogenesis is driven by aberrant self-renewal that is mediated by hyper-active transcriptional machinery, and introduce several molecularly targeted therapies in the making which specifically perturb this transactivation system.


Subject(s)
Leukemia , Myeloid-Lymphoid Leukemia Protein , Carcinogenesis , Hematopoietic Stem Cells , Humans , Leukemia/genetics , Leukemia/therapy , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics
5.
Nucleic Acids Res ; 42(7): 4241-56, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24465000

ABSTRACT

Mixed-lineage leukemia (MLL) maintains the expression of cellular memory genes during development, while leukemic MLL fusion proteins aberrantly maintain expression of hematopoietic stem cell program genes such as HOXA9 to cause leukemia. However, the molecular mechanism of gene activation is unclear. Here we show that only two functional modules are necessary and sufficient for target recognition: those that bind to non-methylated CpGs and di-/tri-methylated histone H3 lysine 36 (H3K36me2/3). An artificial protein composed of the two targeting modules and an interaction domain for AF4-family coactivators can functionally substitute for MLL fusion proteins. Because H3K36me2/3 markers are indicative of active transcription, MLL fusion proteins target previously active CpG-rich genes and activate transcription by recruiting coactivators thereto. Our results indicate that such chromatin context-dependent gene activation is the fundamental mechanism by which MLL fusion proteins maintain the expression of the cellular memory/hematopoietic stem cell program genes.


Subject(s)
CpG Islands , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Histones/metabolism , Humans , Leukemia, Experimental/genetics , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/chemistry , Nucleosomes/metabolism , Oncogene Proteins, Fusion/chemistry , Protein Structure, Tertiary
6.
Rinsho Ketsueki ; 56(10): 1942-9, 2015 Oct.
Article in Japanese | MEDLINE | ID: mdl-26458432

ABSTRACT

Chromosomal translocations at the MLL gene generate chimeric genes of MLL and its partner, thereby causing aggressive acute leukemia. The sequence of the MLL gene was revealed in the early 1990s. Several years later, murine leukemia models using genetic engineering or retroviral gene transfer techniques were established, which spurred basic research on this disease. Since then, a series of technological breakthroughs such as DNA arrays, shRNA library screening, and deep sequencing have provided us with a much deeper understanding of the molecular basis of leukemogenesis caused by MLL mutations. Based on the understanding of these molecular mechanisms, several small molecules that inhibit critical processes of leukemogenesis have been developed as molecularly-targeted drug candidates. I herein review the rapid progress in this research on the molecular mechanisms of MLL-associated leukemia.


Subject(s)
Cell Transformation, Neoplastic , Gene Rearrangement , Leukemia/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Humans , Leukemia/pathology , Protein Binding , Transcription, Genetic
7.
Biochim Biophys Acta Gene Regul Mech ; 1867(3): 195045, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38851533

ABSTRACT

The histone acetyltransferase HBO1, also known as KAT7, is a major chromatin modifying enzyme responsible for H3 and H4 acetylation. It is found within two distinct tetrameric complexes, the JADE subunit-containing complex and BRPF subunit-containing complex. The HBO1-JADE complex acetylates lysine 5, 8 and 12 of histone H4, and the HBO1-BRPF complex acetylates lysine 14 of histone H3. HBO1 regulates gene transcription, DNA replication, DNA damage repair, and centromere function. It is involved in diverse signaling pathways and plays crucial roles in development and stem cell biology. Recent work has established a strong relationship of HBO1 with the histone methyltransferase MLL/KMT2A in acute myeloid leukemia. Here, we discuss functional and pathological links of HBO1 to cancer, highlighting the underlying mechanisms that may pave the way to the development of novel anti-cancer therapies.

8.
Nat Struct Mol Biol ; 31(7): 1039-1049, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38448574

ABSTRACT

JADE is a core subunit of the HBO1 acetyltransferase complex that regulates developmental and epigenetic programs and promotes gene transcription. Here we describe the mechanism by which JADE facilitates recruitment of the HBO1 complex to chromatin and mediates its enzymatic activity. Structural, genomic and complex assembly in vivo studies show that the PZP (PHD1-zinc-knuckle-PHD2) domain of JADE engages the nucleosome through binding to histone H3 and DNA and is necessary for the association with chromatin targets. Recognition of unmethylated H3K4 by PZP directs enzymatic activity of the complex toward histone H4 acetylation, whereas H3K4 hypermethylation alters histone substrate selectivity. We demonstrate that PZP contributes to leukemogenesis, augmenting transforming activity of the NUP98-JADE2 fusion. Our findings highlight biological consequences and the impact of the intact JADE subunit on genomic recruitment, enzymatic function and pathological activity of the HBO1 complex.


Subject(s)
Histone Acetyltransferases , Histones , Humans , Histones/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Animals , Chromatin/metabolism , Acetylation , Mice , Nucleosomes/metabolism , Protein Binding , Methylation , Models, Molecular , Protein Domains , Homeodomain Proteins , Tumor Suppressor Proteins
9.
iScience ; 27(4): 109367, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38500836

ABSTRACT

Acetylation of histones by lysine acetyltransferases (KATs) provides a fundamental mechanism by which chromatin structure and transcriptional programs are regulated. Here, we describe a dual binding activity of the first winged helix domain of human MORF KAT (MORFWH1) that recognizes the TAZ2 domain of p300 KAT (p300TAZ2) and CpG rich DNA sequences. Structural and biochemical studies identified distinct DNA and p300TAZ2 binding sites, allowing MORFWH1 to independently engage either ligand. Genomic data show that MORF/MOZWH1 colocalizes with H3K18ac, a product of enzymatic activity of p300, on CpG rich promoters of target genes. Our findings suggest a functional cooperation of MORF and p300 KATs in transcriptional regulation.

10.
J Cell Sci ; 124(Pt 13): 2208-19, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21670200

ABSTRACT

The mixed lineage leukemia (MLL) proto-oncogenic protein is a histone-lysine N-methyltransferase that is produced by proteolytic cleavage and self-association of the respective functionally distinct subunits (MLL(N) and MLL(C)) to form a holocomplex involved in epigenetic transcriptional regulation. On the basis of studies in Drosophila it has been suggested that the separated subunits might also have distinct functions. In this study, we used a genetically engineered mouse line that lacked MLL(C) to show that the MLL(N)-MLL(C) holocomplex is responsible for MLL functions in various developmental processes. The stability of MLL(N) is dependent on its intramolecular interaction with MLL(C), which is mediated through the first and fourth plant homeodomain (PHD) fingers (PHD1 and PHD4) and the phenylalanine/tyrosine-rich (FYRN) domain of MLL(N). Free MLL(N) is destroyed by a mechanism that targets the FYRN domain, whereas free MLL(C) is exported to the cytoplasm and degraded by the proteasome. PHD1 is encoded by an alternatively spliced exon that is occasionally deleted in T-cell leukemia, and its absence produces an MLL mutant protein that is deficient for holocomplex formation. Therefore, this should be a loss-of-function mutant allele, suggesting that the known tumor suppression role of MLL may also apply to the T-cell lineage. Our data demonstrate that the dissociated MLL subunits are subjected to distinct degradation pathways and thus not likely to have separate functions unless the degradation mechanisms are inhibited.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Leukemia, T-Cell/genetics , Mice , Mice, Knockout , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb-Group Proteins , Protein Processing, Post-Translational , Protein Subunits/genetics , Protein Subunits/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
11.
Cancer Rep (Hoboken) ; 6(2): e1697, 2023 02.
Article in English | MEDLINE | ID: mdl-36806720

ABSTRACT

BACKGROUND: A mathematical model of the radiation-induced cancer was devised to explain the change of incidence rates pursued by Radiation Effect Research Foundation for 25 years. AIM: The aim of this work is construction of mechanisms of radiation-induced cancer and cancers observed in the daily life. METHODS AND RESULTS: First, we found a way to separate spontaneous cancers from radiation-induced cancers observed among atomic-bomb victims in Hiroshima and Nagasaki districts by using a constructed algorithm. The isolated incidence rates of radiation-induced cancers were reproduced by a two-stage model mechanical collision of impinging radiation with cells and succeeding mutation of the damaged cell to cancer. This model satisfactorily reproduced observed solid cancer incidence rates. We further attempted to construct a mathematical model for the age-dependence of spontaneous cancers appearing in the daily life and concluded that the cancer should be generated at cell division. CONCLUSION: With these findings, we reached to a conclusion that cancers may be suppressed by eliminating damaged cells with mild-dose radiation.


Subject(s)
Neoplasms, Radiation-Induced , Nuclear Warfare , Nuclear Weapons , Humans , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Incidence , Models, Theoretical
12.
Nat Commun ; 14(1): 1979, 2023 04 08.
Article in English | MEDLINE | ID: mdl-37031220

ABSTRACT

Changes in the transcriptional machinery cause aberrant self-renewal of non-stem hematopoietic progenitors. AF10 fusions, such as CALM-AF10, are generated via chromosomal translocations, causing malignant leukemia. In this study, we demonstrate that AF10 fusion proteins cause aberrant self-renewal via ENL, which binds to MOZ/MORF lysine acetyltransferases (KATs). The interaction of ENL with MOZ, via its YEATS domain, is critical for CALM-AF10-mediated leukemic transformation. The MOZ/ENL complex recruits DOT1L/AF10 fusion complexes and maintains their chromatin retention via KAT activity. Therefore, inhibitors of MOZ/MORF KATs directly suppress the functions of AF10 fusion proteins, thereby exhibiting strong antitumor effects on AF10 translocation-induced leukemia. Combinatorial inhibition of MOZ/MORF and DOT1L cooperatively induces differentiation of CALM-AF10-leukemia cells. These results reveal roles for the MOZ/ENL complex as an essential recruiting factor of the AF10 fusion/DOT1L complex, providing a rationale for using MOZ/MORF KAT inhibitors in AF10 translocation-induced leukemia.


Subject(s)
Leukemia , Humans , Leukemia/pathology , Myeloid-Lymphoid Leukemia Protein/genetics , Protein Domains , Chromatin , Translocation, Genetic , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism
13.
Cell Death Dis ; 14(9): 597, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37679316

ABSTRACT

Insulin signaling often plays a role in the regulation of cancer, including tumor initiation, progression, and response to treatment. In addition, the insulin-regulated PI3K-Akt-mTOR pathway plays an important role in the regulation of islet cell proliferation, and this pathway is hyperactivated in human non-functional pancreatic neuroendocrine tumors (PanNETs). We, therefore, investigated the effect of a very low carbohydrate diet (ketogenic diet) on a mouse model that develops non-functional PanNETs to ask how reduced PI3K-Akt-mTOR signaling might affect the development and progression of non-functional PanNET. We found that this dietary intervention resulted in lower PI3K-Akt-mTOR signaling in islet cells and a significant reduction in PanNET formation and progression. We also found that this treatment had a significant effect on the suppression of pituitary NET development. Furthermore, we found that non-functional PanNET patients with lower blood glucose levels tend to have a better prognosis than patients with higher blood glucose levels. This preclinical study shows that a dietary intervention that results in lower serum insulin levels leads to lower insulin signals within the neuroendocrine cells and has a striking suppressive effect on the development and progression of both pancreatic and pituitary NETs.


Subject(s)
Neuroendocrine Tumors , Animals , Mice , Humans , Blood Glucose , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Diet, Carbohydrate-Restricted , Insulin , TOR Serine-Threonine Kinases , Receptor Protein-Tyrosine Kinases
14.
Nat Commun ; 14(1): 697, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754959

ABSTRACT

Human acetyltransferases MOZ and MORF are implicated in chromosomal translocations associated with aggressive leukemias. Oncogenic translocations involve the far amino terminus of MOZ/MORF, the function of which remains unclear. Here, we identified and characterized two structured winged helix (WH) domains, WH1 and WH2, in MORF and MOZ. WHs bind DNA in a cooperative manner, with WH1 specifically recognizing unmethylated CpG sequences. Structural and genomic analyses show that the DNA binding function of WHs targets MORF/MOZ to gene promoters, stimulating transcription and H3K23 acetylation, and WH1 recruits oncogenic fusions to HOXA genes that trigger leukemogenesis. Cryo-EM, NMR, mass spectrometry and mutagenesis studies provide mechanistic insight into the DNA-binding mechanism, which includes the association of WH1 with the CpG-containing linker DNA and binding of WH2 to the dyad of the nucleosome. The discovery of WHs in MORF and MOZ and their DNA binding functions could open an avenue in developing therapeutics to treat diseases associated with aberrant MOZ/MORF acetyltransferase activities.


Subject(s)
Acetyltransferases , Histone Acetyltransferases , Leukemia , Humans , Acetylation , Acetyltransferases/metabolism , CpG Islands/genetics , Histone Acetyltransferases/metabolism , Leukemia/genetics , Translocation, Genetic
15.
Nat Commun ; 14(1): 8372, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102116

ABSTRACT

ATP-dependent chromatin remodeling SWI/SNF complexes exist in three subcomplexes: canonical BAF (cBAF), polybromo BAF (PBAF), and a newly described non-canonical BAF (ncBAF). While cBAF and PBAF regulate fates of multiple cell types, roles for ncBAF in hematopoietic stem cells (HSCs) have not been investigated. Motivated by recent discovery of disrupted expression of BRD9, an essential component of ncBAF, in multiple cancers, including clonal hematopoietic disorders, we evaluate here the role of BRD9 in normal and malignant HSCs. BRD9 loss enhances chromatin accessibility, promoting myeloid lineage skewing while impairing B cell development. BRD9 significantly colocalizes with CTCF, whose chromatin recruitment is augmented by BRD9 loss, leading to altered chromatin state and expression of myeloid-related genes within intact topologically associating domains. These data uncover ncBAF as critical for cell fate specification in HSCs via three-dimensional regulation of gene expression and illuminate roles for ncBAF in normal and malignant hematopoiesis.


Subject(s)
Chromatin , Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Cell Differentiation , Hematopoietic Stem Cells/metabolism
16.
FEBS J ; 289(24): 7987-8002, 2022 12.
Article in English | MEDLINE | ID: mdl-34482632

ABSTRACT

Homeostasis in the blood system is maintained by the balance between self-renewing stem cells and nonstem cells. To promote self-renewal, transcriptional regulators maintain epigenetic information during multiple rounds of cell division. Mutations in such transcriptional regulators cause aberrant self-renewal, leading to leukemia. MOZ, a histone acetyltransferase, and MLL, a histone methyltransferase, are transcriptional regulators that promote the self-renewal of hematopoietic stem cells. Gene rearrangements of MOZ and MLL generate chimeric genes encoding fusion proteins that function as constitutively active forms. These MOZ and MLL fusion proteins constitutively activate transcription of their target genes and cause aberrant self-renewal in committed hematopoietic progenitors, which normally do not self-renew. Recent progress in the field suggests that MOZ and MLL are part of a transcriptional activation system that activates the transcription of genes with nonmethylated CpG-rich promoters. The nonmethylated state of CpGs is normally maintained during cell divisions from the mother cell to the daughter cells. Thus, the MOZ/MLL-mediated transcriptional activation system replicates the expression profile of mother cells in daughter cells by activating the transcription of genes previously transcribed in the mother cell. This review summarizes the functions of the components of the MOZ/MLL-mediated transcriptional activation system and their roles in the promotion of self-renewal.


Subject(s)
Leukemia , Humans , Transcriptional Activation , Leukemia/genetics , Leukemia/metabolism , Histone Acetyltransferases/genetics , Histone Acetyltransferases/metabolism , Hematopoietic Stem Cells/metabolism , Carcinogenesis/metabolism , Hematopoiesis/genetics
17.
Nat Commun ; 13(1): 6688, 2022 11 05.
Article in English | MEDLINE | ID: mdl-36335100

ABSTRACT

Chromosomal translocation generates the MLL-AF4 fusion gene, which causes acute leukemia of multiple lineages. MLL-AF4 is a strong oncogenic driver that induces leukemia without additional mutations and is the most common cause of pediatric leukemia. However, establishment of a murine disease model via retroviral transduction has been difficult owning to a lack of understanding of its regulatory mechanisms. Here, we show that MLL-AF4 protein is post-transcriptionally regulated by RNA-binding proteins, including those of KHDRBS and IGF2BP families. MLL-AF4 translation is inhibited by ribosomal stalling, which occurs at regulatory sites containing AU-rich sequences recognized by KHDRBSs. Synonymous mutations disrupting the association of KHDRBSs result in proper translation of MLL-AF4 and leukemic transformation. Consequently, the synonymous MLL-AF4 mutant induces leukemia in vivo. Our results reveal that post-transcriptional regulation critically controls the oncogenic activity of MLL-AF4; these findings might be valuable in developing novel therapies via modulation of the activity of RNA-binding proteins.


Subject(s)
Leukemia , Oncogene Proteins, Fusion , Humans , Mice , Child , Animals , Oncogene Proteins, Fusion/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Translocation, Genetic , Leukemia/genetics , RNA-Binding Proteins/genetics , Oncogenes
18.
Biochim Biophys Acta Mol Cell Res ; 1869(11): 119332, 2022 11.
Article in English | MEDLINE | ID: mdl-35940372

ABSTRACT

Ultraviolet (UV) light irradiation generates pyrimidine dimers on DNA, such as cyclobutane pyrimidine dimers (CPDs) and (6-4) photoproducts. Such dimers distort the high-order DNA structure and prevent transcription and replication. The nucleotide excision repair (NER) system contributes to resolving this type of DNA lesion. There are two pathways that recognize pyrimidine dimers. One acts on transcribed strands of DNA (transcription-coupled NER), and the other acts on the whole genome (global genome-NER; GG-NER). In the latter case, DNA damage-binding protein 2 (DDB2) senses pyrimidine dimers with several histone modification enzymes. We previously reported that histone acetyltransferase binding to ORC1 (HBO1) interacts with DDB2 and facilitates recruitment of the imitation switch chromatin remodeler at UV-irradiated sites via an unknown methyltransferase. Here, we found that the phosphorylated histone methyltransferase mixed lineage leukemia 1 (MLL1) was maintained at UV-irradiated sites in an HBO1-dependent manner. Furthermore, MLL1 catalyzed histone H3K4 methylation and recruited the chromatin remodeler bromodomain adjacent to zinc finger domain 1A (BAZ1A)/ATP-utilizing chromatin assembly and remodeling factor 1 (ACF1). Depletion of MLL1 suppressed BAZ1A accumulation at UV-irradiated sites and inhibited the removal of CPDs. These data indicate that the DDB2-HBO1-MLL1 axis is essential for the recruitment of BAZ1A to facilitate GG-NER.


Subject(s)
Leukemia , Pyrimidine Dimers , Chromatin/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage , DNA Repair , Humans , Pyrimidine Dimers/chemistry , Pyrimidine Dimers/metabolism
19.
Leukemia ; 36(11): 2605-2620, 2022 11.
Article in English | MEDLINE | ID: mdl-36229594

ABSTRACT

Myeloid malignancies with DDX41 mutations are often associated with bone marrow failure and cytopenia before overt disease manifestation. However, the mechanisms underlying these specific conditions remain elusive. Here, we demonstrate that loss of DDX41 function impairs efficient RNA splicing, resulting in DNA replication stress with excess R-loop formation. Mechanistically, DDX41 binds to the 5' splice site (5'SS) of coding RNA and coordinates RNA splicing and transcriptional elongation; loss of DDX41 prevents splicing-coupled transient pausing of RNA polymerase II at 5'SS, causing aberrant R-loop formation and transcription-replication collisions. Although the degree of DNA replication stress acquired in S phase is small, cells undergo mitosis with under-replicated DNA being remained, resulting in micronuclei formation and significant DNA damage, thus leading to impaired cell proliferation and genomic instability. These processes may be responsible for disease phenotypes associated with DDX41 mutations.


Subject(s)
RNA Splice Sites , RNA Splicing , Cell Line , RNA Splicing/genetics , Mutation , DNA Replication
20.
STAR Protoc ; 2(2): 100404, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33855306

ABSTRACT

Conventional chromatin immunoprecipitation (ChIP) includes many steps that need to be optimized. Here, we have described a protocol of fractionation-assisted native ChIP (fanChIP) which combines subfractionation and native ChIP to purify protein/chromatin complexes applicable for analyses of both protein-protein and protein-DNA interactions within a short period of time. fanChIP is advantageous as subcellular fractionation removes chromatin-unbound materials before immunoprecipitation, and the chromatin fragmentation by micrococcal nuclease (MNase) in a mild condition enables one-step purification of intact protein/chromatin complexes. For complete details on the use and execution of this protocol, please refer to Miyamoto et al. (2020).


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin , DNA , Chromatin/genetics , Chromatin/metabolism , DNA/analysis , DNA/chemistry , DNA/metabolism , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Sequence Analysis, DNA , Sequence Analysis, Protein
SELECTION OF CITATIONS
SEARCH DETAIL