Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 133
Filter
1.
Cell ; 170(5): 875-888.e20, 2017 Aug 24.
Article in English | MEDLINE | ID: mdl-28757253

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is one of the most lethal human malignancies, owing in part to its propensity for metastasis. Here, we used an organoid culture system to investigate how transcription and the enhancer landscape become altered during discrete stages of disease progression in a PDA mouse model. This approach revealed that the metastatic transition is accompanied by massive and recurrent alterations in enhancer activity. We implicate the pioneer factor FOXA1 as a driver of enhancer activation in this system, a mechanism that renders PDA cells more invasive and less anchorage-dependent for growth in vitro, as well as more metastatic in vivo. In this context, FOXA1-dependent enhancer reprogramming activates a transcriptional program of embryonic foregut endoderm. Collectively, our study implicates enhancer reprogramming, FOXA1 upregulation, and a retrograde developmental transition in PDA metastasis.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Pancreatic Ductal/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/genetics , Pancreatic Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Disease Models, Animal , Epigenomics , Female , Gene Expression Profiling , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Organoids/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology
2.
Mol Cell ; 84(6): 1149-1157.e7, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38309274

ABSTRACT

OCA-B, OCA-T1, and OCA-T2 belong to a family of coactivators that bind to POU transcription factors (TFs) to regulate gene expression in immune cells. Here, we identify IκBζ (encoded by the NFKBIZ gene) as an additional coactivator of POU TFs. Although originally discovered as an inducible regulator of NF-κB, we show here that IκBζ shares a microhomology with OCA proteins and uses this segment to bind to POU TFs and octamer-motif-containing DNA. Our functional experiments suggest that IκBζ requires its interaction with POU TFs to coactivate immune-related genes. This finding is reinforced by epigenomic analysis of MYD88L265P-mutant lymphoma cells, which revealed colocalization of IκBζ with the POU TF OCT2 and NF-κB:p50 at hundreds of DNA elements harboring octamer and κB motifs. These results suggest that IκBζ is a transcriptional coactivator that can amplify and integrate the output of NF-κB and POU TFs at inducible genes in immune cells.


Subject(s)
DNA , NF-kappa B , NF-kappa B/genetics , NF-kappa B/metabolism , Promoter Regions, Genetic , DNA/genetics , DNA/metabolism
3.
Cell ; 166(3): 536-537, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27471963

ABSTRACT

Identifying molecular biomarkers that predict cancer drug efficacy is crucial for the advancement of precision medicine. In this issue of Cell, Iorio et al. nominate hundreds of potential genetic and epigenetic biomarkers through high-throughput drug screening in ∼1,000 molecularly annotated cancer cell lines.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasms/diagnosis , Antineoplastic Agents , Cell Line , Humans , Precision Medicine
4.
Cell ; 157(4): 869-81, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24813610

ABSTRACT

Fragile X syndrome, a common form of inherited intellectual disability, is caused by loss of the fragile X mental retardation protein FMRP. FMRP is present predominantly in the cytoplasm, where it regulates translation of proteins that are important for synaptic function. We identify FMRP as a chromatin-binding protein that functions in the DNA damage response (DDR). Specifically, we show that FMRP binds chromatin through its tandem Tudor (Agenet) domain in vitro and associates with chromatin in vivo. We also demonstrate that FMRP participates in the DDR in a chromatin-binding-dependent manner. The DDR machinery is known to play important roles in developmental processes such as gametogenesis. We show that FMRP occupies meiotic chromosomes and regulates the dynamics of the DDR machinery during mouse spermatogenesis. These findings suggest that nuclear FMRP regulates genomic stability at the chromatin interface and may impact gametogenesis and some developmental aspects of fragile X syndrome.


Subject(s)
Spermatogenesis , Animals , Chromatin/metabolism , Chromosome Pairing , DNA Damage , Embryo, Mammalian/cytology , Fibroblasts , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Hippocampus/cytology , Histones/metabolism , Humans , Male , Meiosis , Mice , Mice, Knockout , Mutation , Neurons/metabolism , Prophase , Receptors, AMPA/metabolism
5.
Nature ; 613(7942): 195-202, 2023 01.
Article in English | MEDLINE | ID: mdl-36544023

ABSTRACT

Inhibition of the tumour suppressive function of p53 (encoded by TP53) is paramount for cancer development in humans. However, p53 remains unmutated in the majority of cases of glioblastoma (GBM)-the most common and deadly adult brain malignancy1,2. Thus, how p53-mediated tumour suppression is countered in TP53 wild-type (TP53WT) GBM is unknown. Here we describe a GBM-specific epigenetic mechanism in which the chromatin regulator bromodomain-containing protein 8 (BRD8) maintains H2AZ occupancy at p53 target loci through the EP400 histone acetyltransferase complex. This mechanism causes a repressive chromatin state that prevents transactivation by p53 and sustains proliferation. Notably, targeting the bromodomain of BRD8 displaces H2AZ, enhances chromatin accessibility and engages p53 transactivation. This in turn enforces cell cycle arrest and tumour suppression in TP53WT GBM. In line with these findings, BRD8 is highly expressed with H2AZ in proliferating single cells of patient-derived GBM, and is inversely correlated with CDKN1A, a canonical p53 target that encodes p21 (refs. 3,4). This work identifies BRD8 as a selective epigenetic vulnerability for a malignancy for which treatment has not improved for decades. Moreover, targeting the bromodomain of BRD8 may be a promising therapeutic strategy for patients with TP53WT GBM.


Subject(s)
Epigenesis, Genetic , Glioblastoma , Transcription Factors , Tumor Suppressor Protein p53 , Adult , Humans , Cell Cycle Checkpoints , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Histones/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cell Proliferation
6.
Cell ; 153(2): 320-34, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582323

ABSTRACT

Chromatin regulators have become attractive targets for cancer therapy, but it is unclear why inhibition of these ubiquitous regulators should have gene-specific effects in tumor cells. Here, we investigate how inhibition of the widely expressed transcriptional coactivator BRD4 leads to selective inhibition of the MYC oncogene in multiple myeloma (MM). BRD4 and Mediator were found to co-occupy thousands of enhancers associated with active genes. They also co-occupied a small set of exceptionally large super-enhancers associated with genes that feature prominently in MM biology, including the MYC oncogene. Treatment of MM tumor cells with the BET-bromodomain inhibitor JQ1 led to preferential loss of BRD4 at super-enhancers and consequent transcription elongation defects that preferentially impacted genes with super-enhancers, including MYC. Super-enhancers were found at key oncogenic drivers in many other tumor cells. These observations have implications for the discovery of cancer therapeutics directed at components of super-enhancers in diverse tumor types.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Enhancer Elements, Genetic , Mediator Complex/metabolism , Neoplasms/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Triazoles/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Chromatin , Gene Expression Regulation, Neoplastic/drug effects , Genome-Wide Association Study , Humans , Mediator Complex/antagonists & inhibitors , Multiple Myeloma/genetics , Nuclear Proteins/antagonists & inhibitors , Transcription Elongation, Genetic , Transcription Factors/antagonists & inhibitors
7.
Nature ; 607(7917): 169-175, 2022 07.
Article in English | MEDLINE | ID: mdl-35576971

ABSTRACT

Tuft cells are a rare chemosensory lineage that coordinates immune and neural responses to foreign pathogens in mucosal tissues1. Recent studies have also revealed tuft-cell-like human tumours2,3, particularly as a variant of small-cell lung cancer. Both normal and neoplastic tuft cells share a genetic requirement for the transcription factor POU2F3 (refs. 2,4), although the transcriptional mechanisms that generate this cell type are poorly understood. Here we show that binding of POU2F3 to the uncharacterized proteins C11orf53 and COLCA2 (renamed here OCA-T1/POU2AF2 and OCA-T2/POU2AF3, respectively) is critical in the tuft cell lineage. OCA-T1 and OCA-T2 are paralogues of the B-cell-specific coactivator OCA-B; all three proteins are encoded in a gene cluster and contain a conserved peptide that binds to class II POU transcription factors and a DNA octamer motif in a bivalent manner. We demonstrate that binding between POU2F3 and OCA-T1 or OCA-T2 is essential in tuft-cell-like small-cell lung cancer. Moreover, we generated OCA-T1-deficient mice, which are viable but lack tuft cells in several mucosal tissues. These findings reveal that the POU2F3-OCA-T complex is the master regulator of tuft cell identity and a molecular vulnerability of tuft-cell-like small-cell lung cancer.


Subject(s)
Cell Lineage , Lung Neoplasms , Neoplasm Proteins , Octamer Transcription Factors , Small Cell Lung Carcinoma , Animals , Humans , Mice , Lung Neoplasms/pathology , Mucous Membrane/pathology , Multigene Family/genetics , Neoplasm Proteins/metabolism , Nucleotide Motifs , Octamer Transcription Factors/metabolism , POU Domain Factors/metabolism , Small Cell Lung Carcinoma/pathology , Trans-Activators
8.
Cell ; 146(6): 904-17, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21889194

ABSTRACT

MYC contributes to the pathogenesis of a majority of human cancers, yet strategies to modulate the function of the c-Myc oncoprotein do not exist. Toward this objective, we have targeted MYC transcription by interfering with chromatin-dependent signal transduction to RNA polymerase, specifically by inhibiting the acetyl-lysine recognition domains (bromodomains) of putative coactivator proteins implicated in transcriptional initiation and elongation. Using a selective small-molecule bromodomain inhibitor, JQ1, we identify BET bromodomain proteins as regulatory factors for c-Myc. BET inhibition by JQ1 downregulates MYC transcription, followed by genome-wide downregulation of Myc-dependent target genes. In experimental models of multiple myeloma, a Myc-dependent hematologic malignancy, JQ1 produces a potent antiproliferative effect associated with cell-cycle arrest and cellular senescence. Efficacy of JQ1 in three murine models of multiple myeloma establishes the therapeutic rationale for BET bromodomain inhibition in this disease and other malignancies characterized by pathologic activation of c-Myc.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Multiple Myeloma/drug therapy , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Azepines/chemistry , Azepines/pharmacology , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/genetics , Transcriptional Activation/drug effects , Triazoles/chemistry , Triazoles/pharmacology
9.
Mol Cell ; 69(6): 1017-1027.e6, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29526696

ABSTRACT

The lineage-specific transcription factor (TF) MEF2C is often deregulated in leukemia. However, strategies to target this TF have yet to be identified. Here, we used a domain-focused CRISPR screen to reveal an essential role for LKB1 and its Salt-Inducible Kinase effectors (SIK3, in a partially redundant manner with SIK2) to maintain MEF2C function in acute myeloid leukemia (AML). A key phosphorylation substrate of SIK3 in this context is HDAC4, a repressive cofactor of MEF2C. Consequently, targeting of LKB1 or SIK3 diminishes histone acetylation at MEF2C-bound enhancers and deprives leukemia cells of the output of this essential TF. We also found that MEF2C-dependent leukemias are sensitive to on-target chemical inhibition of SIK activity. This study reveals a chemical strategy to block MEF2C function in AML, highlighting how an oncogenic TF can be disabled by targeting of upstream kinases.


Subject(s)
Leukemia, Myeloid, Acute/enzymology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Acetylation , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation , Enhancer Elements, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , HEK293 Cells , Hep G2 Cells , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , K562 Cells , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mice , NIH 3T3 Cells , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , THP-1 Cells , U937 Cells
10.
Proc Natl Acad Sci U S A ; 120(36): e2303859120, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37639593

ABSTRACT

Recurrent chromosomal rearrangements found in rhabdomyosarcoma (RMS) produce the PAX3-FOXO1 fusion protein, which is an oncogenic driver and a dependency in this disease. One important function of PAX3-FOXO1 is to arrest myogenic differentiation, which is linked to the ability of RMS cells to gain an unlimited proliferation potential. Here, we developed a phenotypic screening strategy for identifying factors that collaborate with PAX3-FOXO1 to block myo-differentiation in RMS. Unlike most genes evaluated in our screen, we found that loss of any of the three subunits of the Nuclear Factor Y (NF-Y) complex leads to a myo-differentiation phenotype that resembles the effect of inactivating PAX3-FOXO1. While the transcriptomes of NF-Y- and PAX3-FOXO1-deficient RMS cells bear remarkable similarity to one another, we found that these two transcription factors occupy nonoverlapping sites along the genome: NF-Y preferentially occupies promoters, whereas PAX3-FOXO1 primarily binds to distal enhancers. By integrating multiple functional approaches, we map the PAX3 promoter as the point of intersection between these two regulators. We show that NF-Y occupies CCAAT motifs present upstream of PAX3 to function as a transcriptional activator of PAX3-FOXO1 expression in RMS. These findings reveal a critical upstream role of NF-Y in the oncogenic PAX3-FOXO1 pathway, highlighting how a broadly essential transcription factor can perform tumor-specific roles in governing cellular state.


Subject(s)
Rhabdomyosarcoma , CCAAT-Binding Factor/genetics , Cell Differentiation/genetics , Chromosome Aberrations , Rhabdomyosarcoma/genetics , Transcription Factors
11.
Genes Dev ; 32(13-14): 915-928, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29945888

ABSTRACT

Small cell lung cancer (SCLC) is widely considered to be a tumor of pulmonary neuroendocrine cells; however, a variant form of this disease has been described that lacks neuroendocrine features. Here, we applied domain-focused CRISPR screening to human cancer cell lines to identify the transcription factor (TF) POU2F3 (POU class 2 homeobox 3; also known as SKN-1a/OCT-11) as a powerful dependency in a subset of SCLC lines. An analysis of human SCLC specimens revealed that POU2F3 is expressed exclusively in variant SCLC tumors that lack expression of neuroendocrine markers and instead express markers of a chemosensory lineage known as tuft cells. Using chromatin- and RNA-profiling experiments, we provide evidence that POU2F3 is a master regulator of tuft cell identity in a variant form of SCLC. Moreover, we show that most SCLC tumors can be classified into one of three lineages based on the expression of POU2F3, ASCL1, or NEUROD1. Our CRISPR screens exposed other unique dependencies in POU2F3-expressing SCLC lines, including the lineage TFs SOX9 and ASCL2 and the receptor tyrosine kinase IGF1R (insulin-like growth factor 1 receptor). These data reveal POU2F3 as a cell identity determinant and a dependency in a tuft cell-like variant of SCLC, which may reflect a previously unrecognized cell of origin or a trans-differentiation event in this disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/physiopathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Humans , Lung/pathology , Mice , Receptor, IGF Type 1/metabolism
12.
Mol Cell ; 68(4): 731-744.e9, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149598

ABSTRACT

Bromodomain and extraterminal domain inhibitors (BETi) represent promising therapeutic agents for metastatic melanoma, yet their mechanism of action remains unclear. Here we interrogated the transcriptional effects of BETi and identified AMIGO2, a transmembrane molecule, as a BET target gene essential for melanoma cell survival. AMIGO2 is upregulated in melanoma cells and tissues compared to human melanocytes and nevi, and AMIGO2 silencing in melanoma cells induces G1/S arrest followed by apoptosis. We identified the pseudokinase PTK7 as an AMIGO2 interactor whose function is regulated by AMIGO2. Epigenomic profiling and genome editing revealed that AMIGO2 is regulated by a melanoma-specific BRD2/4-bound promoter and super-enhancer configuration. Upon BETi treatment, BETs are evicted from these regulatory elements, resulting in AMIGO2 silencing and changes in PTK7 proteolytic processing. Collectively, this study uncovers mechanisms underlying the therapeutic effects of BETi in melanoma and reveals the AMIGO2-PTK7 axis as a targetable pathway for metastatic melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Enhancer Elements, Genetic , Melanoma/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Male , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Gut ; 73(6): 941-954, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38262672

ABSTRACT

OBJECTIVE: The optimal therapeutic response in cancer patients is highly dependent upon the differentiation state of their tumours. Pancreatic ductal adenocarcinoma (PDA) is a lethal cancer that harbours distinct phenotypic subtypes with preferential sensitivities to standard therapies. This study aimed to investigate intratumour heterogeneity and plasticity of cancer cell states in PDA in order to reveal cell state-specific regulators. DESIGN: We analysed single-cell expression profiling of mouse PDAs, revealing intratumour heterogeneity and cell plasticity and identified pathways activated in the different cell states. We performed comparative analysis of murine and human expression states and confirmed their phenotypic diversity in specimens by immunolabeling. We assessed the function of phenotypic regulators using mouse models of PDA, organoids, cell lines and orthotopically grafted tumour models. RESULTS: Our expression analysis and immunolabeling analysis show that a mucus production programme regulated by the transcription factor SPDEF is highly active in precancerous lesions and the classical subtype of PDA - the most common differentiation state. SPDEF maintains the classical differentiation and supports PDA transformation in vivo. The SPDEF tumour-promoting function is mediated by its target genes AGR2 and ERN2/IRE1ß that regulate mucus production, and inactivation of the SPDEF programme impairs tumour growth and facilitates subtype interconversion from classical towards basal-like differentiation. CONCLUSIONS: Our findings expand our understanding of the transcriptional programmes active in precancerous lesions and PDAs of classical differentiation, determine the regulators of mucus production as specific vulnerabilities in these cell states and reveal phenotype switching as a response mechanism to inactivation of differentiation states determinants.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Animals , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Mice , Humans , Mucus/metabolism , Mucoproteins/metabolism , Mucoproteins/genetics , Cell Line, Tumor , Cell Differentiation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteins/metabolism , Proteins/genetics , Organoids/pathology , Organoids/metabolism , Cell Plasticity , Gene Expression Regulation, Neoplastic , Disease Models, Animal , Oncogene Proteins
14.
EMBO J ; 38(19): e96659, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31454099

ABSTRACT

Loss of the histone H3.3-specific chaperone component ATRX or its partner DAXX frequently occurs in human cancers that employ alternative lengthening of telomeres (ALT) for chromosomal end protection, yet the underlying mechanism remains unclear. Here, we report that ATRX/DAXX does not serve as an immediate repressive switch for ALT. Instead, ATRX or DAXX depletion gradually induces telomere DNA replication dysfunction that activates not only homology-directed DNA repair responses but also cell cycle checkpoint control. Mechanistically, we demonstrate that this process is contingent on ATRX/DAXX histone chaperone function, independently of telomere length. Combined ATAC-seq and telomere chromatin immunoprecipitation studies reveal that ATRX loss provokes progressive telomere decondensation that culminates in the inception of persistent telomere replication dysfunction. We further show that endogenous telomerase activity cannot overcome telomere dysfunction induced by ATRX loss, leaving telomere repair-based ALT as the only viable mechanism for telomere maintenance during immortalization. Together, these findings implicate ALT activation as an adaptive response to ATRX/DAXX loss-induced telomere replication dysfunction.


Subject(s)
Co-Repressor Proteins/genetics , Molecular Chaperones/genetics , Telomere Homeostasis , Telomere/metabolism , X-linked Nuclear Protein/genetics , Cell Line , DNA Repair , Gene Deletion , HEK293 Cells , Humans , Telomerase/metabolism
15.
Nature ; 552(7683): 126-131, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29186125

ABSTRACT

N6-methyladenosine (m6A) is an abundant internal RNA modification in both coding and non-coding RNAs that is catalysed by the METTL3-METTL14 methyltransferase complex. However, the specific role of these enzymes in cancer is still largely unknown. Here we define a pathway that is specific for METTL3 and is implicated in the maintenance of a leukaemic state. We identify METTL3 as an essential gene for growth of acute myeloid leukaemia cells in two distinct genetic screens. Downregulation of METTL3 results in cell cycle arrest, differentiation of leukaemic cells and failure to establish leukaemia in immunodeficient mice. We show that METTL3, independently of METTL14, associates with chromatin and localizes to the transcriptional start sites of active genes. The vast majority of these genes have the CAATT-box binding protein CEBPZ present at the transcriptional start site, and this is required for recruitment of METTL3 to chromatin. Promoter-bound METTL3 induces m6A modification within the coding region of the associated mRNA transcript, and enhances its translation by relieving ribosome stalling. We show that genes regulated by METTL3 in this way are necessary for acute myeloid leukaemia. Together, these data define METTL3 as a regulator of a chromatin-based pathway that is necessary for maintenance of the leukaemic state and identify this enzyme as a potential therapeutic target for acute myeloid leukaemia.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Methyltransferases/metabolism , Promoter Regions, Genetic/genetics , Protein Biosynthesis , Adenosine/genetics , Adenosine/metabolism , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin/genetics , Chromatin/metabolism , Female , Genes, Neoplasm/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Methyltransferases/chemistry , Methyltransferases/deficiency , Methyltransferases/genetics , Mice , Protein Biosynthesis/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribosomes/metabolism , Transcription Initiation Site
16.
Mol Cell ; 58(6): 1028-39, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25982114

ABSTRACT

The bromodomain and extraterminal (BET) protein BRD4 is a validated drug target in leukemia, yet its regulatory function in this disease is not well understood. Here, we show that BRD4 chromatin occupancy in acute myeloid leukemia closely correlates with the hematopoietic transcription factors (TFs) PU.1, FLI1, ERG, C/EBPα, C/EBPß, and MYB at nucleosome-depleted enhancer and promoter regions. We provide evidence that these TFs, in conjunction with the lysine acetyltransferase activity of p300/CBP, facilitate BRD4 recruitment to their occupied sites to promote transcriptional activation. Chemical inhibition of BET bromodomains was found to suppress the functional output of each hematopoietic TF, thereby interfering with essential lineage-specific transcriptional circuits in this disease. These findings reveal a chromatin-based signaling cascade comprised of hematopoietic TFs, p300/CBP, and BRD4 that supports leukemia maintenance and is suppressed by BET bromodomain inhibition.


Subject(s)
Hematopoietic System/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Acetylation/drug effects , Acute Disease , Animals , Azepines/pharmacology , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Gene Expression Profiling , Histones/metabolism , Humans , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , NIH 3T3 Cells , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Binding/drug effects , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Regulator ERG , Triazoles/pharmacology
17.
Mol Cell ; 60(6): 847-59, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26626481

ABSTRACT

The bromodomain and extraterminal (BET) protein BRD4 is a therapeutic target in acute myeloid leukemia (AML). Here, we demonstrate that the AML maintenance function of BRD4 requires its interaction with NSD3, which belongs to a subfamily of H3K36 methyltransferases. Unexpectedly, AML cells were found to only require a short isoform of NSD3 that lacks the methyltransferase domain. We show that NSD3-short is an adaptor protein that sustains leukemia by linking BRD4 to the CHD8 chromatin remodeler, by using a PWWP chromatin reader module, and by employing an acidic transactivation domain. Genetic targeting of NSD3 or CHD8 mimics the phenotypic and transcriptional effects of BRD4 inhibition. Furthermore, BRD4, NSD3, and CHD8 colocalize across the AML genome, and each is released from super-enhancer regions upon chemical inhibition of BET bromodomains. These findings suggest that BET inhibitors exert therapeutic effects in leukemia by evicting BRD4-NSD3-CHD8 complexes from chromatin to suppress transcription.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Catalytic Domain , Cell Cycle Proteins , Cell Proliferation , Chromatin Assembly and Disassembly , DNA-Binding Proteins/genetics , HL-60 Cells , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/genetics
18.
Proc Natl Acad Sci U S A ; 117(21): 11471-11482, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32385160

ABSTRACT

Lineage plasticity is a prominent feature of pancreatic ductal adenocarcinoma (PDA) cells, which can occur via deregulation of lineage-specifying transcription factors. Here, we show that the zinc finger protein ZBED2 is aberrantly expressed in PDA and alters tumor cell identity in this disease. Unexpectedly, our epigenomic experiments reveal that ZBED2 is a sequence-specific transcriptional repressor of IFN-stimulated genes, which occurs through antagonism of IFN regulatory factor 1 (IRF1)-mediated transcriptional activation at cooccupied promoter elements. Consequently, ZBED2 attenuates the transcriptional output and growth arrest phenotypes downstream of IFN signaling in multiple PDA cell line models. We also found that ZBED2 is preferentially expressed in the squamous molecular subtype of human PDA, in association with inferior patient survival outcomes. Consistent with this observation, we show that ZBED2 can repress the pancreatic progenitor transcriptional program, enhance motility, and promote invasion in PDA cells. Collectively, our findings suggest that high ZBED2 expression is acquired during PDA progression to suppress the IFN response pathway and to promote lineage plasticity in this disease.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , DNA-Binding Proteins/metabolism , Interferon Regulatory Factor-1/metabolism , Pancreatic Neoplasms/pathology , Transcription Factors/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/genetics , Interferon-gamma/pharmacology , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Promoter Regions, Genetic , Survival Analysis , Transcription Factors/genetics
19.
Genes Dev ; 29(3): 250-61, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25644601

ABSTRACT

The mechanisms by which TGF-ß promotes lung adenocarcinoma (ADC) metastasis are largely unknown. Here, we report that in lung ADC cells, TGF-ß potently induces expression of DOCK4, but not other DOCK family members, via the Smad pathway and that DOCK4 induction mediates TGF-ß's prometastatic effects by enhancing tumor cell extravasation. TGF-ß-induced DOCK4 stimulates lung ADC cell protrusion, motility, and invasion without affecting epithelial-to-mesenchymal transition. These processes, which are fundamental to tumor cell extravasation, are driven by DOCK4-mediated Rac1 activation, unveiling a novel link between TGF-ß and Rac1. Thus, our findings uncover the atypical Rac1 activator DOCK4 as a key component of the TGF-ß/Smad pathway that promotes lung ADC cell extravasation and metastasis.


Subject(s)
Adenocarcinoma/physiopathology , GTPase-Activating Proteins/metabolism , Lung Neoplasms/physiopathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Metastasis
20.
Blood ; 135(1): 56-70, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31697837

ABSTRACT

Lineage-defining transcription factors (TFs) are compelling targets for leukemia therapy, yet they are among the most challenging proteins to modulate directly with small molecules. We previously used CRISPR screening to identify a salt-inducible kinase 3 (SIK3) requirement for the growth of acute myeloid leukemia (AML) cell lines that overexpress the lineage TF myocyte enhancer factor (MEF2C). In this context, SIK3 maintains MEF2C function by directly phosphorylating histone deacetylase 4 (HDAC4), a repressive cofactor of MEF2C. In this study, we evaluated whether inhibition of SIK3 with the tool compound YKL-05-099 can suppress MEF2C function and attenuate disease progression in animal models of AML. Genetic targeting of SIK3 or MEF2C selectively suppressed the growth of transformed hematopoietic cells under in vitro and in vivo conditions. Similar phenotypes were obtained when cells were exposed to YKL-05-099, which caused cell-cycle arrest and apoptosis in MEF2C-expressing AML cell lines. An epigenomic analysis revealed that YKL-05-099 rapidly suppressed MEF2C function by altering the phosphorylation state and nuclear localization of HDAC4. Using a gatekeeper allele of SIK3, we found that the antiproliferative effects of YKL-05-099 occurred through on-target inhibition of SIK3 kinase activity. Based on these findings, we treated 2 different mouse models of MLL-AF9 AML with YKL-05-099, which attenuated disease progression in vivo and extended animal survival at well-tolerated doses. These findings validate SIK3 as a therapeutic target in MEF2C-addicted AML and provide a rationale for developing druglike inhibitors of SIK3 for definitive preclinical investigation and for studies in human patients.


Subject(s)
Aniline Compounds/pharmacology , Leukemia, Myeloid, Acute/prevention & control , MEF2 Transcription Factors/metabolism , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Pyrimidines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Apoptosis , Cell Cycle , Cell Proliferation , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , MEF2 Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL