Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cell ; 183(5): 1143-1146, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33128870

ABSTRACT

Given the heterogeneity of senescent cells, our knowledge of both the drivers and consequences of cellular senescence in tissues and organs remains limited, as is our understanding of how this process could be harnessed for human health. Here we identified five broad areas that would help propel the field forward.


Subject(s)
Cellular Senescence , Biomarkers/metabolism , Clinical Trials as Topic , Humans , Models, Biological
2.
Cell ; 165(2): 357-71, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27058666

ABSTRACT

We report a mechanism through which the transcription machinery directly controls topoisomerase 1 (TOP1) activity to adjust DNA topology throughout the transcription cycle. By comparing TOP1 occupancy using chromatin immunoprecipitation sequencing (ChIP-seq) versus TOP1 activity using topoisomerase 1 sequencing (TOP1-seq), a method reported here to map catalytically engaged TOP1, TOP1 bound at promoters was discovered to become fully active only after pause-release. This transition coupled the phosphorylation of the carboxyl-terminal-domain (CTD) of RNA polymerase II (RNAPII) with stimulation of TOP1 above its basal rate, enhancing its processivity. TOP1 stimulation is strongly dependent on the kinase activity of BRD4, a protein that phosphorylates Ser2-CTD and regulates RNAPII pause-release. Thus the coordinated action of BRD4 and TOP1 overcame the torsional stress opposing transcription as RNAPII commenced elongation but preserved negative supercoiling that assists promoter melting at start sites. This nexus between transcription and DNA topology promises to elicit new strategies to intercept pathological gene expression.


Subject(s)
DNA Topoisomerases, Type I/metabolism , DNA/metabolism , RNA Polymerase II/metabolism , Transcription, Genetic , DNA/chemistry , DNA Topoisomerases, Type I/genetics , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , RNA Polymerase II/chemistry , RNA Polymerase II/isolation & purification , Transcription Elongation, Genetic , Transcription Factors/isolation & purification , Transcription Initiation Site
3.
Mol Cell ; 83(3): 335-342, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36640770

ABSTRACT

Genomic architecture appears to play crucial roles in health and a variety of diseases. How nuclear structures reorganize over different timescales is elusive, partly because the tools needed to probe and perturb them are not as advanced as needed by the field. To fill this gap, the National Institutes of Health Common Fund started a program in 2015, called the 4D Nucleome (4DN), with the goal of developing and ultimately applying technologies to interrogate the structure and function of nuclear organization in space and time.


Subject(s)
Cell Nucleus , Genome , United States , Cell Nucleus/genetics , Genomics
4.
Chromosoma ; 133(3): 203-216, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38922437

ABSTRACT

Transgenerational gene expression depends on both underlying DNA sequences and epigenetic modifications. The latter, which can result in transmission of variegated gene expression patterns across multiple generations without DNA alterations, has been termed epigenetic inheritance and has been documented in plants, worms, flies and mammals. Whereas transcription factors binding to cognate DNA sequence elements regulate gene expression, the molecular basis for epigenetic inheritance has been linked to histone and DNA modifications and non-coding RNA. Here we report that mutation of the CCAAT box promoter element abrogates NF-Y binding and disrupts the stable transgenerational expression of an MHC class I transgene. Transgenic mice with a mutated CCAAT box in the MHC class I transgene display variegated expression of the transgene among littermates and progeny in multiple independently derived transgenic lines. After 4 generations, CCAAT mutant transgenic lines derived from a single founder stably displayed distinct patterns of expression. Histone modifications and RNA polymerase II binding correlate with expression of CCAAT mutant transgenic lines, whereas DNA methylation and nucleosome occupancy do not. Mutation of the CCAAT box also results in changes to CTCF binding and DNA looping patterns across the transgene that correlate with expression status. These studies identify the CCAAT promoter element as a regulator of stable transgenerational gene expression such that mutation of the CCAAT box results in variegated transgenerational inheritance. Considering that the CCAAT box is present in 30% of eukaryotic promoters, this study provides insights into how fidelity of gene expression patterns is maintained through multiple generations.


Subject(s)
Mice, Transgenic , Promoter Regions, Genetic , Animals , Mice , DNA Methylation , Epigenesis, Genetic , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Gene Expression Regulation , Genes, MHC Class I , Mutation , Histones/metabolism , Histones/genetics , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Transgenes , Nucleosomes/metabolism , Nucleosomes/genetics
5.
Proc Natl Acad Sci U S A ; 117(24): 13457-13467, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32482868

ABSTRACT

The protooncogene MYC regulates a variety of cellular processes, including proliferation and metabolism. Maintaining MYC at homeostatic levels is critical to normal cell function; overexpression drives many cancers. MYC stability is regulated through phosphorylation: phosphorylation at Thr58 signals degradation while Ser62 phosphorylation leads to its stabilization and functional activation. The bromodomain protein 4 (BRD4) is a transcriptional and epigenetic regulator with intrinsic kinase and histone acetyltransferase (HAT) activities that activates transcription of key protooncogenes, including MYC We report that BRD4 phosphorylates MYC at Thr58, leading to MYC ubiquitination and degradation, thereby regulating MYC target genes. Importantly, BRD4 degradation, but not inhibition, results in increased levels of MYC protein. Conversely, MYC inhibits BRD4's HAT activity, suggesting that MYC regulates its own transcription by limiting BRD4-mediated chromatin remodeling of its locus. The MYC stabilizing kinase, ERK1, regulates MYC levels directly and indirectly by inhibiting BRD4 kinase activity. These findings demonstrate that BRD4 negatively regulates MYC levels, which is counteracted by ERK1 activation.


Subject(s)
Cell Cycle Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/metabolism , Acetylation , Cell Nucleus/metabolism , Chromatin/metabolism , Dipeptides/pharmacology , Gene Expression Regulation/drug effects , HeLa Cells , Heterocyclic Compounds, 3-Ring/pharmacology , Histones/metabolism , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Binding , Protein Stability/drug effects , Proto-Oncogene Proteins c-myc/genetics , Ubiquitination
6.
J Biol Chem ; 297(5): 101326, 2021 11.
Article in English | MEDLINE | ID: mdl-34688663

ABSTRACT

Bromodomain protein 4 (BRD4) is a transcriptional and epigenetic regulator that is a therapeutic target in many cancers and inflammatory diseases. BRD4 plays important roles in transcription as an active kinase, which phosphorylates the carboxy-terminal domain (CTD) of RNA polymerase II (Pol II), the proto-oncogene c-MYC, and transcription factors TAF7 and CDK9. BRD4 is also a passive scaffold that recruits transcription factors. Despite these well-established functions, there has been little characterization of BRD4's biophysical properties or its kinase activity. We report here that the 156 kD mouse BRD4 exists in an extended dimeric conformation with a sedimentation coefficient of ∼6.7 S and a high frictional ratio. Deletion of the conserved B motif (aa 503-548) disrupts BRD4's dimerization. BRD4 kinase activity maps to amino acids 351 to 598, which span bromodomain-2, the B motif, and the BID domain (BD2-B-BID) and contributes to the in vivo phosphorylation of its substrates. As further assessed by analytical ultracentrifugation, BRD4 directly binds purified Pol II CTD. Importantly, the conserved A motif of BRD4 is essential for phosphorylation of Pol II CTD, but not for phosphorylation of TAF7, mapping its binding site to the A motif. Peptides of the viral MLV integrase (MLVIN) protein and cellular histone lysine methyltransferase, NSD3, which have been shown by NMR to bind to the extra-terminal (ET) domain, also are phosphorylated by BRD4. Thus, BRD4 has multiple distinct substrate-binding sites and a common kinase domain. These results provide new insights into the structure and kinase function of BRD4.


Subject(s)
Nuclear Proteins/chemistry , Protein Kinases/chemistry , Protein Multimerization , Transcription Factors/chemistry , Amino Acid Motifs , Animals , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Domains , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Quaternary , RNA Polymerase II/chemistry , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription Factor TFIID/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Immunity ; 38(6): 1116-28, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23746651

ABSTRACT

Immune tolerance requires regulatory T (Treg) cells to prevent autoimmune disease, with the transcription factor Foxp3 functioning as the critical regulator of Treg cell development and function. We report here that Foxp3 was lethal to developing Treg cells in the thymus because it induced a unique proapoptotic protein signature (Puma⁺⁺⁺p-Bim⁺⁺p-JNK⁺⁺DUSP6⁻) and repressed expression of prosurvival Bcl-2 molecules. However, Foxp3 lethality was prevented by common gamma chain (γc)-dependent cytokine signals that were present in the thymus in limiting amounts sufficient to support only ∼1 million Treg cells. Consequently, most newly arising Treg cells in the thymus were deprived of this signal and underwent Foxp3-induced death, with Foxp3⁺CD25⁻ Treg precursor cells being the most susceptible. Thus, we identify Foxp3 as a proapoptotic protein that requires developing Treg cells to compete with one another for limiting amounts of γc-dependent survival signals in the thymus.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cytokines/immunology , Forkhead Transcription Factors/metabolism , Interleukin Receptor Common gamma Subunit/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Survival , Cells, Cultured , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Lymphopoiesis/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
8.
Trends Biochem Sci ; 40(3): 165-71, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25680757

ABSTRACT

Early studies established that transcription initiates within an approximately 50 bp DNA segment capable of nucleating the assembly of RNA polymerase II (Pol II) and associated general transcription factors (GTFs) necessary for transcriptional initiation; this region is called a core promoter. Subsequent analyses identified a series of conserved DNA sequence elements, present in various combinations or not at all, in core promoters. Recent genome-wide analyses have provided further insights into the complexity of core promoter architecture and function. Here we review recent studies that delineate the active role of core promoters in the transcriptional regulation of diverse physiological systems.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic/genetics , Transcription, Genetic , Animals , Genome-Wide Association Study , Humans , RNA Polymerase II/metabolism
9.
J Biol Chem ; 291(1): 342-54, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26504077

ABSTRACT

Most of the steps in, and many of the factors contributing to, glucocorticoid receptor (GR)-regulated gene induction are currently unknown. A competition assay, based on a validated chemical kinetic model of steroid hormone action, is now used to identify two new factors (BRD4 and negative elongation factor (NELF)-E) and to define their sites and mechanisms of action. BRD4 is a kinase involved in numerous initial steps of gene induction. Consistent with its complicated biochemistry, BRD4 is shown to alter both the maximal activity (Amax) and the steroid concentration required for half-maximal induction (EC50) of GR-mediated gene expression by acting at a minimum of three different kinetically defined steps. The action at two of these steps is dependent on BRD4 concentration, whereas the third step requires the association of BRD4 with P-TEFb. BRD4 is also found to bind to NELF-E, a component of the NELF complex. Unexpectedly, NELF-E modifies GR induction in a manner that is independent of the NELF complex. Several of the kinetically defined steps of BRD4 in this study are proposed to be related to its known biochemical actions. However, novel actions of BRD4 and of NELF-E in GR-controlled gene induction have been uncovered. The model-based competition assay is also unique in being able to order, for the first time, the sites of action of the various reaction components: GR < Cdk9 < BRD4 ≤ induced gene < NELF-E. This ability to order factor actions will assist efforts to reduce the side effects of steroid treatments.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Animals , Binding, Competitive , Cell Cycle Proteins , Cyclin-Dependent Kinase 9/metabolism , HeLa Cells , Humans , Kinetics , Mutant Proteins/metabolism , Mutation , Nuclear Receptor Coactivator 2/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding , Rats
10.
J Immunol ; 192(6): 2892-903, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24523508

ABSTRACT

Expression of MHC class I molecules, which provide immune surveillance against intracellular pathogens, is higher on lymphoid cells than on any other cell types. In T cells, this is a result of activation of class I transcription by the T cell enhanceosome consisting of Runx1, CBFß, and LEF1. We now report that MHC class I transcription in T cells also is enhanced by Foxp3, resulting in higher levels of class I in CD4(+)CD25(+) T regulatory cells than in conventional CD4(+)CD25(-) T cells. Interestingly, the effect of Foxp3 regulation of MHC class I transcription is cell type specific: Foxp3 increases MHC class I expression in T cells but represses it in epithelial tumor cells. In both cell types, Foxp3 targets the upstream IFN response element and downstream core promoter of the class I gene. Importantly, expression of MHC class I contributes to the function of CD4(+)CD25(+) T regulatory cells by enhancing immune suppression, both in in vitro and in vivo. These findings identify MHC class I genes as direct targets of Foxp3 whose expression augments regulatory T cell function.


Subject(s)
Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HeLa Cells , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Jurkat Cells , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes, Regulatory/metabolism , Thymocytes/immunology , Thymocytes/metabolism , beta 2-Microglobulin/deficiency , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
11.
Proc Natl Acad Sci U S A ; 109(18): 6927-32, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509028

ABSTRACT

The bromodomain protein, BRD4, has been identified recently as a therapeutic target in acute myeloid leukemia, multiple myeloma, Burkitt's lymphoma, NUT midline carcinoma, colon cancer, and inflammatory disease; its loss is a prognostic signature for metastatic breast cancer. BRD4 also contributes to regulation of both cell cycle and transcription of oncogenes, HIV, and human papilloma virus (HPV). Despite its role in a broad range of biological processes, the precise molecular mechanism of BRD4 function remains unknown. We report that BRD4 is an atypical kinase that binds to the carboxyl-terminal domain (CTD) of RNA polymerase II and directly phosphorylates its serine 2 (Ser2) sites both in vitro and in vivo under conditions where other CTD kinases are inactive. Phosphorylation of the CTD Ser2 is inhibited in vivo by a BRD4 inhibitor that blocks its binding to chromatin. Our finding that BRD4 is an RNA polymerase II CTD Ser2 kinase implicates it as a regulator of eukaryotic transcription.


Subject(s)
Nuclear Proteins/metabolism , RNA Polymerase II/chemistry , RNA Polymerase II/metabolism , Transcription Factors/metabolism , Amino Acid Substitution , Animals , Binding Sites/genetics , Cell Cycle Proteins , Cells, Cultured , Humans , Mice , Mutagenesis, Site-Directed , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphorylation , Protein Structure, Tertiary , RNA Polymerase II/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/chemistry , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic
12.
Biochim Biophys Acta ; 1829(11): 1184-90, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24036077

ABSTRACT

The Major Histocompatibility Complex (MHC) class II transactivator (CIITA) mediates activated immune responses and its deficiency results in the Type II Bare Lymphocyte Syndrome. CIITA is a transcriptional co-activator that regulates γ-interferon-activated transcription of MHC class I and class II genes. It is also a functional homolog of TAF1, a component of the general transcription factor complex TFIID. TAF1 and CIITA both possess intrinsic acetyltransferase (AT) activity that is required for transcription initiation. In response to induction by γ-interferon, CIITA and it's AT activity bypass the requirement for TAF1 AT activity. TAF1 also has kinase activity that is essential for its function. However, no similar activity has been identified for CIITA thus far. Here we report that CIITA, like TAF1, is a serine-threonine kinase. Its substrate specificity parallels, but does not duplicate, that of TAF1 in phosphorylating the TFIID component TAF7, the RAP74 subunit of the general transcription factor TFIIF and histone H2B. Like TAF1, CIITA autophosphorylates, affecting its interaction with TAF7. Additionally, CIITA phosphorylates histone H2B at Ser36, a target of TAF1 that is required for transcription during cell cycle progression and stress response. However, unlike TAF1, CIITA also phosphorylates all the other histones. The identification of this novel kinase activity of CIITA further clarifies its role as a functional homolog of TAF1 which may operate during stress and γ-IFN activated MHC gene transcription.


Subject(s)
Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA Primers , HeLa Cells , Humans , Mass Spectrometry , Phosphorylation , Spodoptera
13.
Cancer Discov ; 14(4): 559-562, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38446429

ABSTRACT

SUMMARY: Cancer Grand Challenges is an international funding initiative that aims to unite the world's best scientists to tackle some of cancer's toughest problems by funding team science on a global scale. Here, we discuss the five newly funded teams and the challenges they will address over the coming years.


Subject(s)
Financing, Organized , Neoplasms , Humans , Neoplasms/therapy
14.
bioRxiv ; 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38352301

ABSTRACT

Oncogene activity rewires cellular transcription, creating new transcription networks to which cancer cells become addicted, by mechanisms that are still poorly understood. Using human and mouse models of T cell acute lymphoblastic leukemia (T-ALL), we identify an essential nuclear role for CHMP5, a cytoplasmic endosomal sorting complex required for transport (ESCRT) protein, in establishing and maintaining the T-ALL transcriptional program. Nuclear CHMP5 promoted the T-ALL gene program by augmenting recruitment of the co-activator BRD4 by the histone acetyl transferase p300 selectively at enhancers and super-enhancers, an interaction that potentiated H3K27 acetylation at these regulatory enhancers. Consequently, loss of CHMP5 diminished BRD4 occupancy at enhancers and super-enhancers and impaired RNA polymerase II pause release, which resulted in downregulation of key T-ALL genes, notably MYC. Reinforcing its importance in T-ALL pathogenesis, CHMP5 deficiency mitigated chemoresistance in human T-ALL cells and abrogated T-ALL induction by oncogenic NOTCH1 in vivo. Thus, the ESCRT protein CHMP5 is an essential positive regulator of the transcriptional machinery promoting T-ALL disease.

15.
J Biol Chem ; 287(46): 38755-66, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23027873

ABSTRACT

The RNA polymerase II (Pol II) C-terminal domain (CTD) serves as a docking site for numerous proteins, bridging various nuclear processes to transcription. The recruitment of these proteins is mediated by CTD phospho-epitopes generated during transcription. The mechanisms regulating the kinases that establish these phosphorylation patterns on the CTD are not known. We report that three CTD kinases, CDK7, CDK9, and BRD4, engage in cross-talk, modulating their subsequent CTD phosphorylation. BRD4 phosphorylates PTEFb/CDK9 at either Thr-29 or Thr-186, depending on its relative abundance, which represses or activates CDK9 CTD kinase activity, respectively. Conversely, CDK9 phosphorylates BRD4 enhancing its CTD kinase activity. The CTD Ser-5 kinase CDK7 also interacts with and phosphorylates BRD4, potently inhibiting BRD4 kinase activity. Additionally, the three kinases regulate each other indirectly through the general transcription factor TAF7. An inhibitor of CDK9 and CDK7 CTD kinase activities, TAF7 also binds to BRD4 and inhibits its kinase activity. Each of these kinases phosphorylates TAF7, affecting its subsequent ability to inhibit the other two. Thus, a complex regulatory network governs Pol II CTD kinases.


Subject(s)
Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinases/chemistry , Nuclear Proteins/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factors/chemistry , Animals , Cell Cycle Proteins , Cell Line , DNA Damage , Drosophila , HeLa Cells , Humans , Models, Biological , Phosphorylation , Protein Structure, Tertiary , RNA Polymerase II/metabolism , Serine/chemistry , Transcription, Genetic , Cyclin-Dependent Kinase-Activating Kinase
16.
Lancet Oncol ; 18(11): e622-e623, 2017 11.
Article in English | MEDLINE | ID: mdl-29208378
17.
Cancer Discov ; 13(4): 819-823, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36884310

ABSTRACT

Cancer Grand Challenges is an international funding initiative that aims to unite the world's best scientists to tackle some of cancer's toughest challenges by funding team science on a global scale. Here, we discuss the nine new, ambitious challenges currently open for application.


Subject(s)
Neoplasms , Humans , Neoplasms/therapy
18.
Sci Adv ; 9(9): eade3876, 2023 03.
Article in English | MEDLINE | ID: mdl-36857449

ABSTRACT

Cohesin, a trimeric complex that establishes sister chromatid cohesion, has additional roles in chromatin organization and transcription. We report that among those roles is the regulation of alternative splicing through direct interactions and in situ colocalization with splicing factors. Degradation of cohesin results in marked changes in splicing, independent of its effects on transcription. Introduction of a single cohesin point mutation in embryonic stem cells alters splicing patterns, demonstrating causality. In primary human acute myeloid leukemia, mutations in cohesin are highly correlated with distinct patterns of alternative splicing. Cohesin also directly interacts with BRD4, another splicing regulator, to generate a pattern of splicing that is distinct from either factor alone, documenting their functional interaction. These findings identify a role for cohesin in regulating alternative splicing in both normal and leukemic cells and provide insights into the role of cohesin mutations in human disease.


Subject(s)
Alternative Splicing , Nuclear Proteins , Humans , Transcription Factors , Cell Cycle Proteins , Cohesins
19.
bioRxiv ; 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37333336

ABSTRACT

Transgenerational epigenetic inheritance is defined as the transmission of traits or gene expression patterns across multiple generations that do not derive from DNA alterations. The effect of multiple stress factors or metabolic changes resulting in such inheritance have been documented in plants, worms and flies and mammals. The molecular basis for epigenetic inheritance has been linked to histone and DNA modifications and non-coding RNA. In this study, we show that mutation of a promoter element, the CCAAT box, disrupts stable expression of an MHC Class I transgene, resulting in variegated expression among progeny for at least 4 generations in multiple independently derived transgenic lines. Histone modifications and RNA polII binding correlate with expression, whereas DNA methylation and nucleosome occupancy do not. Mutation of the CCAAT box abrogates NF-Y binding and results in changes to CTCF binding and DNA looping patterns across the gene that correlate with expression status from one generation to the next. These studies identify the CCAAT promoter element as a regulator of stable transgenerational epigenetic inheritance. Considering that the CCAAT box is present in 30% of eukaryotic promoters, this study could provide important insights into how fidelity of gene expression patterns is maintained through multiple generations.

20.
Mol Cancer Res ; 21(2): 91-101, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36378541

ABSTRACT

Small-cell lung cancer (SCLC) is a recalcitrant malignancy that urgently needs new therapies. Four master transcription factors (ASCL1, NEUROD1, POU2F3, and YAP1) have been identified in SCLC, and each defines the transcriptome landscape of one molecular subtype. However, these master transcription factors have not been found directly druggable. We hypothesized that blocking their transcriptional coactivator(s) could provide an alternative approach to target these master transcription factors. Here, we identify that BET proteins physically interact with NEUROD1 and function as transcriptional coactivators. Using CRISPR knockout and ChIP-seq, we demonstrate that NEUROD1 plays a critical role in defining the landscapes of BET proteins in the SCLC genome. Blocking BET proteins by inhibitors led to broad suppression of the NEUROD1-target genes, especially those associated with superenhancers, resulting in the inhibition of SCLC growth in vitro and in vivo. LSAMP, a membrane protein in the IgLON family, was identified as one of the NEUROD1-target genes mediating BET inhibitor sensitivity in SCLC. Altogether, our study reveals that BET proteins are essential in regulating NEUROD1 transactivation and are promising targets in SCLC-N subtype tumors. IMPLICATIONS: Our findings suggest that targeting transcriptional coactivators could be a novel approach to blocking the master transcription factors in SCLC for therapeutic purposes.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Transcriptional Activation , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL