Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Harm Reduct J ; 21(1): 46, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38378660

ABSTRACT

BACKGROUND: Xylazine is a dangerous veterinary sedative found mainly in illicit fentanyl in the Northeast and Midwest. Its role in the Deep South overdose crisis is not well-characterized. METHODS: We conducted a retrospective review of autopsy data in Jefferson County, Alabama to identify trends in xylazine prevalence among people who fatally overdosed from June 2019 through June 2023. RESULTS: 165 decedents met inclusion criteria. While the first identified xylazine-associated overdose was in June 2019, xylazine has become consistently prevalent since January 2021. All cases of xylazine-associated fatal overdoses were accompanied by fentanyl, and most (75.4%) involved poly-drug stimulant use. The average age was 42.2, and most decedents were white (58.8%) and male (68.5%). Overall, 18.2% of people were unhoused at the time of death. DISCUSSION: Xylazine is prevalent in the Deep South. Efforts to promote harm reduction, publicly viewable drug supply trends, and legalization of drug checking and syringe service programs should be prioritized.


Subject(s)
Drug Overdose , Illicit Drugs , Humans , Male , Adult , Fentanyl , Analgesics, Opioid , Retrospective Studies , Xylazine , Drug Overdose/epidemiology
3.
Clin Cancer Res ; 27(20): 5669-5680, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34433651

ABSTRACT

PURPOSE: The epigenetic mechanisms involved in transcriptional regulation leading to malignant phenotype in gliomas remains poorly understood. Topoisomerase IIB (TOP2B), an enzyme that decoils and releases torsional forces in DNA, is overexpressed in a subset of gliomas. Therefore, we investigated its role in epigenetic regulation in these tumors. EXPERIMENTAL DESIGN: To investigate the role of TOP2B in epigenetic regulation in gliomas, we performed paired chromatin immunoprecipitation sequencing for TOP2B and RNA-sequencing analysis of glioma cell lines with and without TOP2B inhibition and in human glioma specimens. These experiments were complemented with assay for transposase-accessible chromatin using sequencing, gene silencing, and mouse xenograft experiments to investigate the function of TOP2B and its role in glioma phenotypes. RESULTS: We discovered that TOP2B modulates transcription of multiple oncogenes in human gliomas. TOP2B regulated transcription only at sites where it was enzymatically active, but not at all native binding sites. In particular, TOP2B activity localized in enhancers, promoters, and introns of PDGFRA and MYC, facilitating their expression. TOP2B levels and genomic localization was associated with PDGFRA and MYC expression across glioma specimens, which was not seen in nontumoral human brain tissue. In vivo, TOP2B knockdown of human glioma intracranial implants prolonged survival and downregulated PDGFRA. CONCLUSIONS: Our results indicate that TOP2B activity exerts a pleiotropic role in transcriptional regulation of oncogenes in a subset of gliomas promoting a proliferative phenotype.


Subject(s)
Brain Neoplasms/genetics , DNA Topoisomerases, Type II/physiology , Epigenesis, Genetic/physiology , Glioma/genetics , Introns/physiology , Oncogenes/physiology , Poly-ADP-Ribose Binding Proteins/physiology , Promoter Regions, Genetic/physiology , Animals , Brain Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Glioma/enzymology , Humans , Mice
4.
Proc Natl Acad Sci U S A ; 117(44): 27365-27373, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33077595

ABSTRACT

Actively transcribed genes in mammals are decorated by H3K79 methylation, which is correlated with transcription levels and is catalyzed by the histone methyltransferase DOT1L. DOT1L is required for mammalian development, and the inhibition of its catalytic activity has been extensively studied for cancer therapy; however, the mechanisms underlying DOT1L's functions in normal development and cancer pathogenesis remain elusive. To dissect the relationship between H3K79 methylation, cellular differentiation, and transcription regulation, we systematically examined the role of DOT1L and its catalytic activity in embryonic stem cells (ESCs). DOT1L is dispensable for ESC self-renewal but is required for establishing the proper expression signature of neural progenitor cells, while catalytic inactivation of DOT1L has a lesser effect. Furthermore, DOT1L loss, rather than its catalytic inactivation, causes defects in glial cell specification. Although DOT1L loss by itself has no major defect in transcription elongation, transcription elongation defects seen with the super elongation complex inhibitor KL-2 are exacerbated in DOT1L knockout cells, but not in catalytically dead DOT1L cells, revealing a role of DOT1L in promoting productive transcription elongation that is independent of H3K79 methylation. Taken together, our study reveals a catalytic-independent role of DOT1L in modulating cell-fate determination and in transcriptional elongation control.


Subject(s)
Cell Differentiation/genetics , Histone-Lysine N-Methyltransferase/metabolism , Transcription Elongation, Genetic/physiology , Cell Proliferation/drug effects , DNA Methylation/physiology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/genetics , Epigenomics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Humans , Lysine/metabolism , Methylation , Methyltransferases/metabolism , Neural Stem Cells/metabolism , Protein Processing, Post-Translational , Transcriptional Elongation Factors/metabolism
5.
Sci Adv ; 6(26): eaaz4764, 2020 06.
Article in English | MEDLINE | ID: mdl-32637595

ABSTRACT

Set1A and Set1B, two members of the COMPASS family of methyltransferases that methylate the histone H3 lysine 4 (H3K4) residue, have been accredited as primary depositors of global H3K4 trimethylation (H3K4me3) in mammalian cells. Our previous studies in mouse embryonic stem cells (ESCs) demonstrated that deleting the enzymatic SET domain of Set1A does not perturb bulk H3K4me3, indicating possible compensatory roles played by other COMPASS methyltransferases. Here, we generated a series of ESC lines harboring compounding mutations of COMPASS methyltransferases. We find that Set1B is functionally redundant to Set1A in implementing H3K4me3 at highly expressed genes, while Mll2 deposits H3K4me3 at less transcriptionally active promoters. While Set1A-B/COMPASS is responsible for broad H3K4me3 peaks, Mll2/COMPASS establishes H3K4me3 with narrow breadth. Additionally, Mll2 helps preserve global H3K4me3 levels and peak breadth in the absence of Set1A-B activity. Our results illustrate the biological flexibility of such enzymes in regulating transcription in a context-dependent manner to maintain stem cell identity.

6.
Nat Genet ; 52(6): 615-625, 2020 06.
Article in English | MEDLINE | ID: mdl-32393859

ABSTRACT

The COMPASS protein family catalyzes histone H3 Lys 4 (H3K4) methylation and its members are essential for regulating gene expression. MLL2/COMPASS methylates H3K4 on many developmental genes and bivalent clusters. To understand MLL2-dependent transcriptional regulation, we performed a CRISPR-based screen with an MLL2-dependent gene as a reporter in mouse embryonic stem cells. We found that MLL2 functions in gene expression by protecting developmental genes from repression via repelling PRC2 and DNA methylation machineries. Accordingly, repression in the absence of MLL2 is relieved by inhibition of PRC2 and DNA methyltransferases. Furthermore, DNA demethylation on such loci leads to reactivation of MLL2-dependent genes not only by removing DNA methylation but also by opening up previously CpG methylated regions for PRC2 recruitment, diluting PRC2 at Polycomb-repressed genes. These findings reveal how the context and function of these three epigenetic modifiers of chromatin can orchestrate transcriptional decisions and demonstrate that prevention of active repression by the context of the enzyme and not H3K4 trimethylation underlies transcriptional regulation on MLL2/COMPASS targets.


Subject(s)
DNA Methylation , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Animals , Chromosomal Proteins, Non-Histone/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Lysine/metabolism , Methylation , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/physiology , Myeloid-Lymphoid Leukemia Protein/genetics , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics
7.
Mol Cell ; 78(1): 112-126.e12, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243828

ABSTRACT

Delineating how chromosomes fold at length scales beyond one megabase remains obscure relative to smaller-scale folding into TADs, loops, and nucleosomes. We find that rather than simply unfolding chromatin, histone hyperacetylation results in interactions between distant genomic loci separated by tens to hundreds of megabases, even in the absence of transcription. These hyperacetylated "megadomains" are formed by the BRD4-NUT fusion oncoprotein, interact both within and between chromosomes, and form a specific nuclear subcompartment that has elevated gene activity with respect to other subcompartments. Pharmacological degradation of BRD4-NUT results in collapse of megadomains and attenuation of the interactions between them. In contrast, these interactions persist and contacts between newly acetylated regions are formed after inhibiting RNA polymerase II initiation. Our structure-function approach thus reveals that broad chromatin domains of identical biochemical composition, independent of transcription, form nuclear subcompartments, and also indicates the potential of altering chromosome structure for treating human disease.


Subject(s)
Cell Nucleus/genetics , Chromatin/metabolism , Chromosomes, Mammalian/chemistry , Acetylation , Cell Line , Cell Nucleus/metabolism , Chromatin/chemistry , Chromatin/drug effects , Chromosomes, Mammalian/metabolism , Gene Expression , Humans , Male , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/metabolism
8.
Mol Cell ; 78(2): 261-274.e5, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32155413

ABSTRACT

RNA polymerase II (RNA Pol II) is generally paused at promoter-proximal regions in most metazoans, and based on in vitro studies, this function has been attributed to the negative elongation factor (NELF). Here, we show that upon rapid depletion of NELF, RNA Pol II fails to be released into gene bodies, stopping instead around the +1 nucleosomal dyad-associated region. The transition to the 2nd pause region is independent of positive transcription elongation factor P-TEFb. During the heat shock response, RNA Pol II is rapidly released from pausing at heat shock-induced genes, while most genes are paused and transcriptionally downregulated. Both of these aspects of the heat shock response remain intact upon NELF loss. We find that NELF depletion results in global loss of cap-binding complex from chromatin without global reduction of nascent transcript 5' cap stability. Thus, our studies implicate NELF functioning in early elongation complexes distinct from RNA Pol II pause-release.


Subject(s)
Positive Transcriptional Elongation Factor B/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Heat-Shock Response/genetics , Humans , Mice , Nucleosomes/genetics , Promoter Regions, Genetic
9.
Sci Adv ; 5(7): eaax0698, 2019 07.
Article in English | MEDLINE | ID: mdl-31392276

ABSTRACT

The zinc finger of the cerebellum (ZIC) proteins has been implicated to function in normal tissue development. Recent studies have described the critical functions of Zic proteins in cancers and the potential tumor-suppressive functions in colon cancer development and progression. To elucidate the functional roles of Zic proteins in colorectal cancer, we knocked out the Zic5 gene and analyzed the chromatin localization pattern and transcriptional regulation of target gene expression. We found that Zic5 regulates glucose metabolism, and Zic5 knockout is accompanied by an increased glycolytic state and tolerance to a low-glucose condition. Furthermore, loss of ß-catenin or TCF7l2 diminishes the chromatin binding of Zic5 globally. Our studies suggest that the Wnt/ß-catenin signaling pathway has a strong influence on the function of Zic proteins and glucose metabolism in colorectal cancers through GLUT1. Interfering Wnt/-catenin-Zic5 axis-regulated aerobic glycolysis represents a potentially effective strategy to selectively target colon cancer cells.


Subject(s)
Colonic Neoplasms/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glucose Transporter Type 1/genetics , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factors/metabolism , beta Catenin/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Glucose/pharmacology , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Humans , Repressor Proteins/metabolism , Up-Regulation/drug effects
10.
Crit Care Med ; 47(11): 1539-1548, 2019 11.
Article in English | MEDLINE | ID: mdl-31393323

ABSTRACT

OBJECTIVES: To characterize emergency department sedation practices in mechanically ventilated patients, and test the hypothesis that deep sedation in the emergency department is associated with worse outcomes. DESIGN: Multicenter, prospective cohort study. SETTING: The emergency department and ICUs of 15 medical centers. PATIENTS: Mechanically ventilated adult emergency department patients. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: All data involving sedation (medications, monitoring) were recorded. Deep sedation was defined as Richmond Agitation-Sedation Scale of -3 to -5 or Sedation-Agitation Scale of 2 or 1. A total of 324 patients were studied. Emergency department deep sedation was observed in 171 patients (52.8%), and was associated with a higher frequency of deep sedation in the ICU on day 1 (53.8% vs 20.3%; p < 0.001) and day 2 (33.3% vs 16.9%; p = 0.001), when compared to light sedation. Mean (SD) ventilator-free days were 18.1 (10.8) in the emergency department deep sedation group compared to 20.0 (9.8) in the light sedation group (mean difference, 1.9; 95% CI, -0.40 to 4.13). Similar results according to emergency department sedation depth existed for ICU-free days (mean difference, 1.6; 95% CI, -0.54 to 3.83) and hospital-free days (mean difference, 2.3; 95% CI, 0.26-4.32). Mortality was 21.1% in the deep sedation group and 17.0% in the light sedation group (between-group difference, 4.1%; odds ratio, 1.30; 0.74-2.28). The occurrence rate of acute brain dysfunction (delirium and coma) was 68.4% in the deep sedation group and 55.6% in the light sedation group (between-group difference, 12.8%; odds ratio, 1.73; 1.10-2.73). CONCLUSIONS: Early deep sedation in the emergency department is common, carries over into the ICU, and may be associated with worse outcomes. Sedation practice in the emergency department and its association with clinical outcomes is in need of further investigation.


Subject(s)
Deep Sedation/statistics & numerical data , Emergency Service, Hospital , Hypnotics and Sedatives/therapeutic use , Intensive Care Units , Respiration, Artificial/statistics & numerical data , Cohort Studies , Coma/epidemiology , Deep Sedation/mortality , Delirium/epidemiology , Female , Hospital Mortality , Humans , Male , Middle Aged , Severity of Illness Index , United States/epidemiology
11.
Clin Cancer Res ; 25(1): 222-239, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30224337

ABSTRACT

PURPOSE: T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disease, affecting children and adults. Chemotherapy treatments show high response rates but have debilitating effects and carry risk of relapse. Previous work implicated NOTCH1 and other oncogenes. However, direct inhibition of these pathways affects healthy tissues and cancer alike. Our goal in this work has been to identify enzymes active in T-ALL whose activity could be targeted for therapeutic purposes. EXPERIMENTAL DESIGN: To identify and characterize new NOTCH1 druggable partners in T-ALL, we coupled studies of the NOTCH1 interactome to expression analysis and a series of functional analyses in cell lines, patient samples, and xenograft models. RESULTS: We demonstrate that ubiquitin-specific protease 7 (USP7) interacts with NOTCH1 and controls leukemia growth by stabilizing the levels of NOTCH1 and JMJD3 histone demethylase. USP7 is highly expressed in T-ALL and is transcriptionally regulated by NOTCH1. In turn, USP7 controls NOTCH1 levels through deubiquitination. USP7 binds oncogenic targets and controls gene expression through stabilization of NOTCH1 and JMJD3 and ultimately H3K27me3 changes. We also show that USP7 and NOTCH1 bind T-ALL superenhancers, and inhibition of USP7 leads to a decrease of the transcriptional levels of NOTCH1 targets and significantly blocks T-ALL cell growth in vitro and in vivo. CONCLUSIONS: These results provide a new model for USP7 deubiquitinase activity through recruitment to oncogenic chromatin loci and regulation of both oncogenic transcription factors and chromatin marks to promote leukemia. Our studies also show that targeting USP7 inhibition could be a therapeutic strategy in aggressive leukemia.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/genetics , Leukemia, T-Cell/genetics , Receptor, Notch1/genetics , Ubiquitin-Specific Peptidase 7/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genetic Therapy , Humans , Jurkat Cells , Leukemia, T-Cell/pathology , Leukemia, T-Cell/therapy , Mice , Signal Transduction/genetics , Xenograft Model Antitumor Assays
12.
Genes Dev ; 33(1-2): 61-74, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30573454

ABSTRACT

Chromosomal translocations of the Mixed-lineage leukemia 1 (MLL1) gene generate MLL chimeras that drive the pathogenesis of acute myeloid and lymphoid leukemia. The untranslocated MLL1 is a substrate for proteolytic cleavage by the endopeptidase threonine aspartase 1 (taspase1); however, the biological significance of MLL1 cleavage by this endopeptidase remains unclear. Here, we demonstrate that taspase1-dependent cleavage of MLL1 results in the destabilization of MLL. Upon loss of taspase1, MLL1 association with chromatin is markedly increased due to the stabilization of its unprocessed version, and this stabilization of the uncleaved MLL1 can result in the displacement of MLL chimeras from chromatin in leukemic cells. Casein kinase II (CKII) phosphorylates MLL1 proximal to the taspase1 cleavage site, facilitating its cleavage, and pharmacological inhibition of CKII blocks taspase1-dependent MLL1 processing, increases MLL1 stability, and results in the displacement of the MLL chimeras from chromatin. Accordingly, inhibition of CKII in a MLL-AF9 mouse model of leukemia delayed leukemic progression in vivo. This study provides insights into the direct regulation of the stability of MLL1 through its cleavage by taspase1, which can be harnessed for targeted therapeutic approaches for the treatment of aggressive leukemia as the result of MLL translocations.


Subject(s)
Endopeptidases/metabolism , Leukemia/therapy , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Chromatin/metabolism , Disease Models, Animal , Disease Progression , Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , Humans , Leukemia/enzymology , Leukemia/genetics , MCF-7 Cells , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Stability , Survival Analysis
13.
Am J Respir Crit Care Med ; 199(12): 1517-1536, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30554520

ABSTRACT

Rationale: The contributions of diverse cell populations in the human lung to pulmonary fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal changes within individual cell populations during pulmonary fibrosis that are important for disease pathogenesis. Objectives: To determine whether single-cell RNA sequencing can reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other cell types in lung tissue from subjects with pulmonary fibrosis compared with control subjects. Methods: We performed single-cell RNA sequencing on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and on one bronchoscopic cryobiospy sample from a patient with idiopathic pulmonary fibrosis. We validated these data using in situ RNA hybridization, immunohistochemistry, and bulk RNA-sequencing on flow-sorted cells from 22 additional subjects. Measurements and Main Results: We identified a distinct, novel population of profibrotic alveolar macrophages exclusively in patients with fibrosis. Within epithelial cells, the expression of genes involved in Wnt secretion and response was restricted to nonoverlapping cells. We identified rare cell populations including airway stem cells and senescent cells emerging during pulmonary fibrosis. We developed a web-based tool to explore these data. Conclusions: We generated a single-cell atlas of pulmonary fibrosis. Using this atlas, we demonstrated heterogeneity within alveolar macrophages and epithelial cells from subjects with pulmonary fibrosis. These results support the feasibility of discovery-based approaches using next-generation sequencing technologies to identify signaling pathways for targeting in the development of personalized therapies for patients with pulmonary fibrosis.


Subject(s)
Cells, Cultured/pathology , Epithelial Cells/pathology , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Sequence Analysis, RNA , Stem Cells/pathology , Transcriptome , Animals , Disease Models, Animal , Female , Humans , Male
14.
Sci Adv ; 4(11): eaau6986, 2018 11.
Article in English | MEDLINE | ID: mdl-30417100

ABSTRACT

The tet methylcytosine dioxygenase 2 (TET2) enzyme catalyzes the conversion of the modified DNA base 5-methylcytosine to 5-hydroxymethylcytosine. TET2 is frequently mutated or dysregulated in multiple human cancers, and loss of TET2 is associated with changes in DNA methylation patterns. Here, using newly developed TET2-specific antibodies and the estrogen response as a model system for studying the regulation of gene expression, we demonstrate that endogenous TET2 occupies active enhancers and facilitates the proper recruitment of estrogen receptor α (ERα). Knockout of TET2 by CRISPR-CAS9 leads to a global increase of DNA methylation at enhancers, resulting in attenuation of the estrogen response. We further identified a positive feedback loop between TET2 and ERα, which further requires MLL3 COMPASS at these enhancers. Together, this study reveals an epigenetic axis coordinating a transcriptional program through enhancer activation via DNA demethylation.


Subject(s)
Breast Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Demethylation , Enhancer Elements, Genetic , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Differentiation , Cohort Studies , DNA Methylation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Dioxygenases , Epigenesis, Genetic , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Female , Humans , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Survival Rate , Tumor Cells, Cultured
15.
Cancer Cell ; 34(5): 707-723.e7, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30423293

ABSTRACT

CHAF1B is the p60 subunit of the chromatin assembly factor (CAF1) complex, which is responsible for assembly of histones H3.1/H4 heterodimers at the replication fork during S phase. Here we report that CHAF1B is required for normal hematopoiesis while its overexpression promotes leukemia. CHAF1B has a pro-leukemia effect by binding chromatin at discrete sites and interfering with occupancy of transcription factors that promote myeloid differentiation, such as CEBPA. Reducing Chaf1b activity by either heterozygous deletion or overexpression of a CAF1 dominant negative allele is sufficient to suppress leukemogenesis in vivo without impairing normal hematopoiesis.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin Assembly Factor-1/metabolism , Chromatin/metabolism , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/pathology , Nucleosomes/metabolism , Proteins/metabolism , Adult , Animals , Binding Sites/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/genetics , Chromatin Assembly Factor-1/genetics , Exoribonucleases , Female , Hematopoiesis/genetics , Humans , Jurkat Cells , Leukemia, Myeloid, Acute/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/physiology , Proteins/genetics , Repressor Proteins , Ribonucleases
16.
Cell ; 175(3): 766-779.e17, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30340042

ABSTRACT

The super elongation complex (SEC) is required for robust and productive transcription through release of RNA polymerase II (Pol II) with its P-TEFb module and promoting transcriptional processivity with its ELL2 subunit. Malfunction of SEC contributes to multiple human diseases including cancer. Here, we identify peptidomimetic lead compounds, KL-1 and its structural homolog KL-2, which disrupt the interaction between the SEC scaffolding protein AFF4 and P-TEFb, resulting in impaired release of Pol II from promoter-proximal pause sites and a reduced average rate of processive transcription elongation. SEC is required for induction of heat-shock genes and treating cells with KL-1 and KL-2 attenuates the heat-shock response from Drosophila to human. SEC inhibition downregulates MYC and MYC-dependent transcriptional programs in mammalian cells and delays tumor progression in a mouse xenograft model of MYC-driven cancer, indicating that small-molecule disruptors of SEC could be used for targeted therapy of MYC-induced cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms, Experimental/drug therapy , Positive Transcriptional Elongation Factor B/metabolism , Repressor Proteins/metabolism , Transcription Elongation, Genetic/drug effects , Transcriptional Elongation Factors/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Drosophila , Female , HCT116 Cells , HEK293 Cells , Heat-Shock Response , Humans , Male , Mice , Mice, Inbred BALB C , Protein Binding/drug effects , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
17.
Nat Med ; 24(6): 758-769, 2018 06.
Article in English | MEDLINE | ID: mdl-29785026

ABSTRACT

The lysine methyltransferase KMT2C (also known as MLL3), a subunit of the COMPASS complex, implements monomethylation of Lys4 on histone H3 (H3K4) at gene enhancers. KMT2C (hereafter referred to as MLL3) frequently incurs point mutations across a range of human tumor types, but precisely how these lesions alter MLL3 function and contribute to oncogenesis is unclear. Here we report a cancer mutational hotspot in MLL3 within the region encoding its plant homeodomain (PHD) repeats and demonstrate that this domain mediates association of MLL3 with the histone H2A deubiquitinase and tumor suppressor BAP1. Cancer-associated mutations in the sequence encoding the MLL3 PHD repeats disrupt the interaction between MLL3 and BAP1 and correlate with poor patient survival. Cancer cells that had PHD-associated MLL3 mutations or lacked BAP1 showed reduced recruitment of MLL3 and the H3K27 demethylase KDM6A (also known as UTX) to gene enhancers. As a result, inhibition of the H3K27 methyltransferase activity of the Polycomb repressive complex 2 (PRC2) in tumor cells harboring BAP1 or MLL3 mutations restored normal gene expression patterns and impaired cell proliferation in vivo. This study provides mechanistic insight into the oncogenic effects of PHD-associated mutations in MLL3 and suggests that restoration of a balanced state of Polycomb-COMPASS activity may have therapeutic efficacy in tumors that bear mutations in the genes encoding these epigenetic factors.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Epigenesis, Genetic , Polycomb-Group Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromatin/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Histone Demethylases/metabolism , Mice, Nude , Mutation/genetics , Nuclear Proteins/metabolism , PHD Zinc Fingers , Protein Binding , Survival Analysis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
18.
Sci Adv ; 4(1): eaap8747, 2018 01.
Article in English | MEDLINE | ID: mdl-29404406

ABSTRACT

Chromatin regulators control cellular differentiation by orchestrating dynamic developmental gene expression programs, and hence, malfunctions in the regulation of chromatin state contribute to both developmental disorders and disease state. Mll4 (Kmt2d), a member of the COMPASS (COMplex of Proteins ASsociated with Set1) protein family that implements histone H3 lysine 4 monomethylation (H3K4me1) at enhancers, is essential for embryonic development and functions as a pancancer tumor suppressor. We define the roles of Mll4/COMPASS and its catalytic activity in the maintenance and exit of ground-state pluripotency in murine embryonic stem cells (ESCs). Mll4 is required for ESC to exit the naive pluripotent state; however, its intrinsic catalytic activity is dispensable for this process. The depletion of the H3K4 demethylase Lsd1 (Kdm1a) restores the ability of Mll4 null ESCs to transition from naive to primed pluripotency. Thus, we define an opposing regulatory axis, wherein Lsd1 and associated co-repressors directly repress Mll4-activated gene targets. This finding has broad reaching implications for human developmental syndromes and the treatment of tumors carrying Mll4 mutations.


Subject(s)
Cell Differentiation , Enhancer Elements, Genetic , Epigenesis, Genetic , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Mouse Embryonic Stem Cells/metabolism , Multiprotein Complexes/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Self Renewal/genetics , Histone-Lysine N-Methyltransferase/chemistry , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Transcription, Genetic
19.
Methods Mol Biol ; 1712: 19-26, 2018.
Article in English | MEDLINE | ID: mdl-29224065

ABSTRACT

A complete understanding of transcription and co-transcriptional RNA processing events by polymerase requires precise and robust approaches to visualize polymerase progress and quantify nascent transcripts on a genome-wide scale. Here, we present a transcriptome-wide method to measure the level of nascent transcribing RNA in a fast and unbiased manner.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , RNA/genetics , Sequence Analysis, RNA , Transcription, Genetic , Animals , DNA/chemistry , DNA/genetics , Humans , RNA/chemistry , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Software , Transcriptome/genetics
20.
Genes Dev ; 31(20): 2056-2066, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29138278

ABSTRACT

Mutations and translocations within the COMPASS (complex of proteins associated with Set1) family of histone lysine methyltransferases are associated with a large number of human diseases, including cancer. Here we report that SET1B/COMPASS, which is essential for cell survival, surprisingly has a cytoplasmic variant. SET1B, but not its SET domain, is critical for maintaining cell viability, indicating a novel catalytic-independent role of SET1B/COMPASS. Loss of SET1B or its unique cytoplasmic-interacting protein, BOD1, leads to up-regulation of expression of numerous genes modulating fatty acid metabolism, including ADIPOR1 (adiponectin receptor 1), COX7C, SDC4, and COQ7 Our detailed molecular studies identify ADIPOR1 signaling, which is inactivated in both obesity and human cancers, as a key target of SET1B/COMPASS. Collectively, our study reveals a cytoplasmic function for a member of the COMPASS family, which could be harnessed for therapeutic regulation of signaling in human diseases, including cancer.


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Histone-Lysine N-Methyltransferase/physiology , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytoplasm/enzymology , Cytoplasm/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , MCF-7 Cells , Mice , Mice, Nude , PR-SET Domains , Protein Subunits/metabolism , Receptors, Adiponectin/metabolism , Signal Transduction , Triple Negative Breast Neoplasms/etiology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...