Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
PLoS Biol ; 21(9): e3002317, 2023 09.
Article in English | MEDLINE | ID: mdl-37747887

ABSTRACT

Translational control is critical for cell fate transitions during development, lineage specification, and tumorigenesis. Here, we show that the transcription factor double homeobox protein 4 (DUX4), and its previously characterized transcriptional program, broadly regulates translation to change the cellular proteome. DUX4 is a key regulator of zygotic genome activation in human embryos, whereas misexpression of DUX4 causes facioscapulohumeral muscular dystrophy (FSHD) and is associated with MHC-I suppression and immune evasion in cancer. We report that translation initiation and elongation factors are disrupted downstream of DUX4 expression in human myoblasts. Genome-wide translation profiling identified mRNAs susceptible to DUX4-induced translation inhibition, including those encoding antigen presentation factors and muscle lineage proteins, while DUX4-induced mRNAs were robustly translated. Endogenous expression of DUX4 in human FSHD myotubes and cancer cell lines also correlated with reduced protein synthesis and MHC-I presentation. Our findings reveal that DUX4 orchestrates cell state conversion by suppressing the cellular proteome while maintaining translation of DUX4-induced mRNAs to promote an early developmental program.


Subject(s)
Homeodomain Proteins , Muscular Dystrophy, Facioscapulohumeral , Transcription Factors , Humans , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Proteome/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nucleic Acids Res ; 52(6): e34, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38375914

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is a network of pathways that degrades transcripts that undergo premature translation termination. In mammals, NMD can be divided into the exon junction complex (EJC)-enhanced and EJC-independent branches. Fluorescence- and luminescence-based reporters have long been effective tools to investigate NMD, yet existing reporters largely focus on the EJC-enhanced pathway. Here, we present a system of reporters for comparative studies of EJC-independent and EJC-enhanced NMD. This system also enables the study of NMD-associated outcomes such as premature termination codon (PTC) readthrough and truncated protein degradation. These reporters are compatible with fluorescence or luminescence-based readouts via transient transfection or stable integration. Using this reporter system, we show that EJC-enhanced NMD RNA levels are reduced by 2- or 9-fold and protein levels are reduced by 7- or 12-fold compared to EJC-independent NMD, depending on the reporter gene used. Additionally, the extent of readthrough induced by G418 and an NMD inhibitor (SMG1i), alone and in combination, varies across NMD substrates. When combined, G418 and SMG1i increase readthrough product levels in an additive manner for EJC-independent reporters, while EJC-enhanced reporters show a synergistic effect. We present these reporters as a valuable toolkit to deepen our understanding of NMD and its associated mechanisms.


Subject(s)
Exons , Genes, Reporter , Genetic Techniques , Nonsense Mediated mRNA Decay , Exons/genetics , Nonsense Mediated mRNA Decay/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , HEK293 Cells , Genes, Reporter/genetics
3.
Br J Cancer ; 128(9): 1723-1732, 2023 05.
Article in English | MEDLINE | ID: mdl-36807337

ABSTRACT

BACKGROUND: A non-invasive endometrial cancer detection tool that can accurately triage symptomatic women for definitive testing would improve patient care. Urine is an attractive biofluid for cancer detection due to its simplicity and ease of collection. The aim of this study was to identify urine-based proteomic signatures that can discriminate endometrial cancer patients from symptomatic controls. METHODS: This was a prospective case-control study of symptomatic post-menopausal women (50 cancers, 54 controls). Voided self-collected urine samples were processed for mass spectrometry and run using sequential window acquisition of all theoretical mass spectra (SWATH-MS). Machine learning techniques were used to identify important discriminatory proteins, which were subsequently combined in multi-marker panels using logistic regression. RESULTS: The top discriminatory proteins individually showed moderate accuracy (AUC > 0.70) for endometrial cancer detection. However, algorithms combining the most discriminatory proteins performed well with AUCs > 0.90. The best performing diagnostic model was a 10-marker panel combining SPRR1B, CRNN, CALML3, TXN, FABP5, C1RL, MMP9, ECM1, S100A7 and CFI and predicted endometrial cancer with an AUC of 0.92 (0.96-0.97). Urine-based protein signatures showed good accuracy for the detection of early-stage cancers (AUC 0.92 (0.86-0.9)). CONCLUSION: A patient-friendly, urine-based test could offer a non-invasive endometrial cancer detection tool in symptomatic women. Validation in a larger independent cohort is warranted.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Humans , Female , Case-Control Studies , Proteomics/methods , Biomarkers , Mass Spectrometry/methods , Endometrial Neoplasms/diagnosis , Fatty Acid-Binding Proteins , Extracellular Matrix Proteins
4.
EMBO J ; 38(21): e100847, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31433507

ABSTRACT

Phosphorylation is a key regulator of protein function under (patho)physiological conditions, and defining site-specific phosphorylation is essential to understand basic and disease biology. In vertebrates, the investigative focus has primarily been on serine, threonine and tyrosine phosphorylation, but mounting evidence suggests that phosphorylation of other "non-canonical" amino acids also regulates critical aspects of cell biology. However, standard methods of phosphoprotein characterisation are largely unsuitable for the analysis of non-canonical phosphorylation due to their relative instability under acidic conditions and/or elevated temperature. Consequently, the complete landscape of phosphorylation remains unexplored. Here, we report an unbiased phosphopeptide enrichment strategy based on strong anion exchange (SAX) chromatography (UPAX), which permits identification of histidine (His), arginine (Arg), lysine (Lys), aspartate (Asp), glutamate (Glu) and cysteine (Cys) phosphorylation sites on human proteins by mass spectrometry-based phosphoproteomics. Remarkably, under basal conditions, and having accounted for false site localisation probabilities, the number of unique non-canonical phosphosites is approximately one-third of the number of observed canonical phosphosites. Our resource reveals the previously unappreciated diversity of protein phosphorylation in human cells, and opens up avenues for high-throughput exploration of non-canonical phosphorylation in all organisms.


Subject(s)
Anions/chemistry , Chromatography, Ion Exchange/methods , Phosphopeptides/analysis , Phosphoproteins/analysis , Proteome/analysis , Computational Biology , HeLa Cells , Humans , Mass Spectrometry , Phosphorylation
5.
Biochem J ; 479(19): 2063-2086, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36240066

ABSTRACT

Previously, we discovered that deletion of c-Rel in the Eµ-Myc mouse model of lymphoma results in earlier onset of disease, a finding that contrasted with the expected function of this NF-κB subunit in B-cell malignancies. Here we report that Eµ-Myc/cRel-/- cells have an unexpected and major defect in the CHK1 pathway. Total and phospho proteomic analysis revealed that Eµ-Myc/cRel-/- lymphomas highly resemble wild-type (WT) Eµ-Myc lymphomas treated with an acute dose of the CHK1 inhibitor (CHK1i) CCT244747. Further analysis demonstrated that this is a consequence of Eµ-Myc/cRel-/- lymphomas having lost expression of CHK1 protein itself, an effect that also results in resistance to CCT244747 treatment in vivo. Similar down-regulation of CHK1 protein levels was also seen in CHK1i resistant U2OS osteosarcoma and Huh7 hepatocellular carcinoma cells. Further investigation revealed that the deubiquitinase USP1 regulates CHK1 proteolytic degradation and that its down-regulation in our model systems is responsible, at least in part, for these effects. We demonstrate that treating WT Eµ-Myc lymphoma cells with the USP1 inhibitor ML323 was highly effective at reducing tumour burden in vivo. Targeting USP1 activity may thus be an alternative therapeutic strategy in MYC-driven tumours.


Subject(s)
Lymphoma , Proto-Oncogene Proteins c-myc , Aminopyridines , Animals , Deubiquitinating Enzymes , Lymphoma/metabolism , Lymphoma/pathology , Mice , NF-kappa B/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines
6.
Biochem J ; 479(19): 2131-2151, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36240067

ABSTRACT

The development of resistance and the activation of bypass pathway signalling represents a major problem for the clinical application of protein kinase inhibitors. While investigating the effect of either a c-Rel deletion or RelAT505A phosphosite knockin on the Eµ-Myc mouse model of B-cell lymphoma, we discovered that both NF-κB subunit mutations resulted in CHK1 inhibitor resistance, arising from either loss or alteration of CHK1 activity, respectively. However, since Eµ-Myc lymphomas depend on CHK1 activity to cope with high levels of DNA replication stress and consequent genomic instability, it was not clear how these mutant NF-κB subunit lymphomas were able to survive. To understand these survival mechanisms and to identify potential compensatory bypass signalling pathways in these lymphomas, we applied a multi-omics strategy. With c-Rel-/- Eµ-Myc lymphomas we observed high levels of Phosphatidyl-inositol 3-kinase (PI3K) and AKT pathway activation. Moreover, treatment with the PI3K inhibitor Pictilisib (GDC-0941) selectively inhibited the growth of reimplanted c-Rel-/- and RelAT505A, but not wild type (WT) Eµ-Myc lymphomas. We also observed up-regulation of a RHO/RAC pathway gene expression signature in both Eµ-Myc NF-κB subunit mutation models. Further investigation demonstrated activation of the RHO/RAC effector p21-activated kinase (PAK) 2. Here, the PAK inhibitor, PF-3758309 successfully overcame resistance of RelAT505A but not WT lymphomas. These findings demonstrate that up-regulation of multiple bypass pathways occurs in CHK1 inhibitor resistant Eµ-Myc lymphomas. Consequently, drugs targeting these pathways could potentially be used as either second line or combinatorial therapies to aid the successful clinical application of CHK1 inhibitors.


Subject(s)
Lymphoma , Phosphatidylinositol 3-Kinases , Animals , Inositol , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Transgenic , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation , p21-Activated Kinases/genetics
7.
Hum Mol Genet ; 29(6): 1030-1043, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32083293

ABSTRACT

Advances in understanding the pathophysiology of facioscapulohumeral dystrophy (FSHD) have led to the discovery of candidate therapeutics, and it is important to identify markers of disease activity to inform clinical trial design. For drugs that inhibit DUX4 expression, measuring DUX4 or DUX4-target gene expression might be an interim measure of drug activity; however, only a subset of FHSD muscle biopsies shows evidence of DUX4 expression. Our prior study showed that MRI T2-STIR-positive muscles had a higher probability of showing DUX4 expression than muscles with normal MRI characteristics. In the current study, we performed a 1-year follow-up assessment of the same muscle with repeat MRI and muscle biopsy. There was little change in MRI characteristics over the 1-year period and, similar to the initial evaluation, MRI T2-STIR-postive muscles had a higher expression of DUX4-regulated genes, as well as genes associated with inflammation, extracellular matrix and cell cycle. Compared to the initial evaluation, overall the level of expression in these gene categories remained stable over the 1-year period; however, there was some variability for each individual muscle biopsied. The pooled data from both the initial and 1-year follow-up evaluations identified several FSHD subgroups based on gene expression, as well as a set of genes-composed of DUX4-target genes, inflammatory and immune genes and cell cycle control genes-that distinguished all of the FSHD samples from the controls. These candidate markers of disease activity need to be replicated in independent datasets and, if validated, may provide useful measures of disease progression and response to therapy.


Subject(s)
Biomarkers/analysis , Gene Expression Regulation , Homeodomain Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , RNA-Seq/methods , Adult , Aged , Case-Control Studies , Female , Humans , Longitudinal Studies , Male , Middle Aged , Muscle, Skeletal/metabolism , Young Adult
8.
Biochem J ; 478(3): 533-551, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33438746

ABSTRACT

Different types of DNA damage can initiate phosphorylation-mediated signalling cascades that result in stimulus specific pro- or anti-apoptotic cellular responses. Amongst its many roles, the NF-κB transcription factor RelA is central to these DNA damage response pathways. However, we still lack understanding of the co-ordinated signalling mechanisms that permit different DNA damaging agents to induce distinct cellular outcomes through RelA. Here, we use label-free quantitative phosphoproteomics to examine the temporal effects of exposure of U2OS cells to either etoposide (ETO) or hydroxyurea (HU) by monitoring the phosphorylation status of RelA and its protein binding partners. Although few stimulus-specific differences were identified in the constituents of phosphorylated RelA interactome after exposure to these DNA damaging agents, we observed subtle, but significant, changes in their phosphorylation states, as a function of both type and duration of treatment. The DNA double strand break (DSB)-inducing ETO invoked more rapid, sustained responses than HU, with regulated targets primarily involved in transcription, cell division and canonical DSB repair. Kinase substrate prediction of ETO-regulated phosphosites suggest abrogation of CDK and ERK1 signalling, in addition to the known induction of ATM/ATR. In contrast, HU-induced replicative stress mediated temporally dynamic regulation, with phosphorylated RelA binding partners having roles in rRNA/mRNA processing and translational initiation, many of which contained a 14-3-3ε binding motif, and were putative substrates of the dual specificity kinase CLK1. Our data thus point to differential regulation of key cellular processes and the involvement of distinct signalling pathways in modulating DNA damage-specific functions of RelA.


Subject(s)
DNA Damage , Protein Processing, Post-Translational , Transcription Factor RelA/physiology , Amino Acid Motifs , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis/physiology , Bone Neoplasms/pathology , Cell Line, Tumor , Chromatography, Liquid , Consensus Sequence , DNA Breaks, Double-Stranded , DNA Replication , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Etoposide/pharmacology , Humans , Hydroxyurea/pharmacology , Osteosarcoma/pathology , Phosphorylation , Protein Interaction Maps , Protein Kinases/metabolism , Proteomics/methods , Tandem Mass Spectrometry , Time Factors
9.
Hum Mol Genet ; 28(23): 3997-4011, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31630170

ABSTRACT

The DUX4 transcription factor is normally expressed in the cleavage-stage embryo and regulates genes involved in embryonic genome activation. Misexpression of DUX4 in skeletal muscle, however, is toxic and causes facioscapulohumeral muscular dystrophy (FSHD). We recently showed DUX4-induced toxicity is due, in part, to the activation of the double-stranded RNA (dsRNA) response pathway and the accumulation of intranuclear dsRNA foci. Here, we determined the composition of DUX4-induced dsRNAs. We found that a subset of DUX4-induced dsRNAs originate from inverted Alu repeats embedded within the introns of DUX4-induced transcripts and from DUX4-induced dsRNA-forming intergenic transcripts enriched for endogenous retroviruses, Alu and LINE-1 elements. However, these repeat classes were also represented in dsRNAs from cells not expressing DUX4. In contrast, pericentric human satellite II (HSATII) repeats formed a class of dsRNA specific to the DUX4 expressing cells. Further investigation revealed that DUX4 can initiate the bidirectional transcription of normally heterochromatin-silenced HSATII repeats. DUX4-induced HSATII RNAs co-localized with DUX4-induced nuclear dsRNA foci and with intranuclear aggregation of EIF4A3 and ADAR1. Finally, gapmer-mediated knockdown of HSATII transcripts depleted DUX4-induced intranuclear ribonucleoprotein aggregates and decreased DUX4-induced cell death, suggesting that HSATII-formed dsRNAs contribute to DUX4 toxicity.


Subject(s)
DNA, Satellite/genetics , Homeodomain Proteins/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Cell Line , DNA, Satellite/metabolism , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Introns , Models, Biological , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Myoblasts/metabolism , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Transcription Factors/genetics
10.
Hum Mol Genet ; 28(3): 476-486, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30312408

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is a common, dominantly inherited disease caused by the epigenetic de-repression of the DUX4 gene, a transcription factor normally repressed in skeletal muscle. As targeted therapies are now possible in FSHD, a better understanding of the relationship between DUX4 activity, muscle pathology and muscle magnetic resonance imaging (MRI) changes is crucial both to understand disease mechanisms and for the design of future clinical trials. Here, we performed MRIs of the lower extremities in 36 individuals with FSHD, followed by needle muscle biopsies in safely accessible muscles. We examined the correlation between MRI characteristics, muscle pathology and expression of DUX4 target genes. Results show that the presence of elevated MRI short tau inversion recovery signal has substantial predictive value in identifying muscles with active disease as determined by histopathology and DUX4 target gene expression. In addition, DUX4 target gene expression was detected only in FSHD-affected muscles and not in control muscles. These results support the use of MRI to identify FSHD muscles most likely to have active disease and higher levels of DUX4 target gene expression and might be useful in early phase therapeutic trials to demonstrate target engagement in therapies aiming to suppress DUX4 expression.


Subject(s)
Homeodomain Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Facioscapulohumeral/diagnostic imaging , Adult , Aged , Biopsy , Female , Gene Expression , Homeodomain Proteins/biosynthesis , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Transcription Factors/genetics
11.
Genes Dev ; 27(24): 2648-62, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24285714

ABSTRACT

Cancer cells frequently depend on chromatin regulatory activities to maintain a malignant phenotype. Here, we show that leukemia cells require the mammalian SWI/SNF chromatin remodeling complex for their survival and aberrant self-renewal potential. While Brg1, an ATPase subunit of SWI/SNF, is known to suppress tumor formation in several cell types, we found that leukemia cells instead rely on Brg1 to support their oncogenic transcriptional program, which includes Myc as one of its key targets. To account for this context-specific function, we identify a cluster of lineage-specific enhancers located 1.7 Mb downstream from Myc that are occupied by SWI/SNF as well as the BET protein Brd4. Brg1 is required at these distal elements to maintain transcription factor occupancy and for long-range chromatin looping interactions with the Myc promoter. Notably, these distal Myc enhancers coincide with a region that is focally amplified in ∼3% of acute myeloid leukemias. Together, these findings define a leukemia maintenance function for SWI/SNF that is linked to enhancer-mediated gene regulation, providing general insights into how cancer cells exploit transcriptional coactivators to maintain oncogenic gene expression programs.


Subject(s)
DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/physiopathology , Proto-Oncogene Proteins c-myc/genetics , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Knockdown Techniques , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , Transcription Factors/genetics
12.
Hum Mol Genet ; 27(R2): R153-R162, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29718206

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is the third most prevalent muscular dystrophy. A progressive disease, it presents clinically as weakness and wasting of the face, shoulder and upper arm muscles, with later involvement of the trunk and lower extremities. FSHD develops through complex genetic and epigenetic events that converge on a common mechanism of toxicity with mis-expression of the transcription factor double homeobox 4 (DUX4). There is currently no treatment available for FSHD. However, the consensus that ectopic DUX4 expression in skeletal muscle is the root cause of FSHD pathophysiology has allowed research efforts to turn toward cultivating a deeper understanding of DUX4 biology and the pathways that underlie FSHD muscle pathology, and to translational studies aimed at developing targeted therapeutics using ever more sophisticated cell and animal-based models of FSHD. This review summarizes recent advances in our understanding of FSHD, including the regulation and activity of DUX4 in its normal developmental roles as well as its pathological contexts. We highlight how these advances raise new questions and challenges for the field as it moves into the next decade of FSHD research.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Muscular Dystrophy, Facioscapulohumeral/physiopathology , Animals , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/embryology , Muscular Dystrophy, Facioscapulohumeral/genetics
13.
PLoS Genet ; 13(3): e1006658, 2017 03.
Article in English | MEDLINE | ID: mdl-28273136

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is caused by the mis-expression of DUX4 in skeletal muscle cells. DUX4 is a transcription factor that activates genes normally associated with stem cell biology and its mis-expression in FSHD cells results in apoptosis. To identify genes and pathways necessary for DUX4-mediated apoptosis, we performed an siRNA screen in an RD rhabdomyosarcoma cell line with an inducible DUX4 transgene. Our screen identified components of the MYC-mediated apoptotic pathway and the double-stranded RNA (dsRNA) innate immune response pathway as mediators of DUX4-induced apoptosis. Further investigation revealed that DUX4 expression led to increased MYC mRNA, accumulation of nuclear dsRNA foci, and activation of the dsRNA response pathway in both RD cells and human myoblasts. Nuclear dsRNA foci were associated with aggregation of the exon junction complex component EIF4A3. The elevation of MYC mRNA, dsRNA accumulation, and EIF4A3 nuclear aggregates in FSHD muscle cells suggest that these processes might contribute to FSHD pathophysiology.


Subject(s)
Apoptosis , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/pathology , Proto-Oncogene Proteins c-myc/genetics , RNA, Double-Stranded/genetics , Rhabdomyosarcoma/genetics , Caspases/metabolism , Cell Death , Cell Line , Cell Survival , DEAD-box RNA Helicases/genetics , Eukaryotic Initiation Factor-4A/genetics , Exons , Gene Expression Regulation , Humans , Immunity, Innate , Mutation , Myoblasts/metabolism , RNA, Double-Stranded/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/genetics
14.
J Biol Chem ; 293(19): 7160-7175, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29567837

ABSTRACT

Members of the bromodomain and extra-terminal domain (BET) family of proteins (bromodomain-containing (BRD) 2, 3, 4, and T) are widely expressed and highly conserved regulators of gene expression in eukaryotes. These proteins have been intimately linked to human disease, and more than a dozen clinical trials are currently underway to test BET-protein inhibitors as modulators of cancer. However, although it is clear that these proteins use their bromodomains to bind both histones and transcription factors bearing acetylated lysine residues, the molecular mechanisms by which BET family proteins regulate gene expression are not well defined. In particular, the functions of the other domains such as the ET domain have been less extensively studied. Here, we examine the properties of the ET domain of BRD3 as a protein/protein interaction module. Using a combination of pulldown and biophysical assays, we demonstrate that BRD3 binds to a range of chromatin-remodeling complexes, including the NuRD, BAF, and INO80 complexes, via a short linear "KIKL" motif in one of the complex subunits. NMR-based structural analysis revealed that, surprisingly, this mode of interaction is shared by the AF9 and ENL transcriptional coregulators that contain an acetyl-lysine-binding YEATS domain and regulate transcriptional elongation. This observation establishes a functional commonality between these two families of cancer-related transcriptional regulators. In summary, our data provide insight into the mechanisms by which BET family proteins might link chromatin acetylation to transcriptional outcomes and uncover an unexpected functional similarity between BET and YEATS family proteins.


Subject(s)
Chromatin Assembly and Disassembly , Peptides/chemistry , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , ATPases Associated with Diverse Cellular Activities , Acetylation , Amino Acid Motifs , Amino Acid Sequence , Biophysical Phenomena , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks , HEK293 Cells , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Nuclear Proteins/metabolism , Protein Binding , Protein Domains , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/physiology , Sequence Homology, Amino Acid , Trans-Activators/chemistry , Transcription Factors
15.
J Pharmacol Exp Ther ; 370(2): 219-230, 2019 08.
Article in English | MEDLINE | ID: mdl-31189728

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is characterized by misexpression of the double homeobox 4 (DUX4) developmental transcription factor in mature skeletal muscle, where it is responsible for muscle degeneration. Preventing expression of DUX4 mRNA is a disease-modifying therapeutic strategy with the potential to halt or reverse the course of disease. We previously reported that agonists of the ß-2 adrenergic receptor suppress DUX4 expression by activating adenylate cyclase to increase cAMP levels. Efforts to further explore this signaling pathway led to the identification of p38 mitogen-activated protein kinase as a major regulator of DUX4 expression. In vitro experiments demonstrate that clinically advanced p38 inhibitors suppress DUX4 expression in FSHD type 1 and 2 myoblasts and differentiating myocytes in vitro with exquisite potency. Individual small interfering RNA-mediated knockdown of either p38α or p38ß suppresses DUX4 expression, demonstrating that each kinase isoform plays a distinct requisite role in activating DUX4 Finally, p38 inhibitors effectively suppress DUX4 expression in a mouse xenograft model of human FSHD gene regulation. These data support the repurposing of existing clinical p38 inhibitors as potential therapeutics for FSHD. The surprise finding that p38α and p38ß isoforms each independently contribute to DUX4 expression offers a unique opportunity to explore the utility of p38 isoform-selective inhibitors to balance efficacy and safety in skeletal muscle. We propose p38 inhibition as a disease-modifying therapeutic strategy for FSHD. SIGNIFICANCE STATEMENT: Facioscapulohumeral muscular dystrophy (FSHD) currently has no treatment options. This work provides evidence that repurposing a clinically advanced p38 inhibitor may provide the first disease-modifying drug for FSHD by suppressing toxic DUX4 expression, the root cause of muscle degeneration in this disease.


Subject(s)
Gene Expression Regulation/drug effects , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/drug therapy , Protein Kinase Inhibitors/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Disease Models, Animal , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Muscular Dystrophy, Facioscapulohumeral/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Protein Kinase Inhibitors/therapeutic use
17.
Blood ; 125(18): 2825-34, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25696920

ABSTRACT

Inhibitors of bromodomain and extraterminal motif proteins (BETs) are being evaluated for the treatment of cancer and other diseases, yet much remains to be learned about how BET proteins function during normal physiology. We used genomic and genetic approaches to examine BET function in a hematopoietic maturation system driven by GATA1, an acetylated transcription factor previously shown to interact with BETs. We found that BRD2, BRD3, and BRD4 were variably recruited to GATA1-regulated genes, with BRD3 binding the greatest number of GATA1-occupied sites. Pharmacologic BET inhibition impaired GATA1-mediated transcriptional activation, but not repression, genome-wide. Mechanistically, BETs promoted chromatin occupancy of GATA1 and subsequently supported transcriptional activation. Using a combination of CRISPR-Cas9-mediated genomic engineering and shRNA approaches, we observed that depletion of either BRD2 or BRD4 alone blunted erythroid gene activation. Surprisingly, depletion of BRD3 only affected erythroid transcription in the context of BRD2 deficiency. Consistent with functional overlap among BET proteins, forced BRD3 expression substantially rescued defects caused by BRD2 deficiency. These results suggest that pharmacologic BET inhibition should be interpreted in the context of distinct steps in transcriptional activation and overlapping functions among BET family members.


Subject(s)
Erythroid Cells/metabolism , Gene Expression Regulation , Hematopoiesis/genetics , Protein Serine-Threonine Kinases/physiology , RNA-Binding Proteins/physiology , Animals , Cells, Cultured , GATA1 Transcription Factor/genetics , Gene Knockdown Techniques , Humans , Mice , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Tertiary , RNA-Binding Proteins/chemistry
18.
Biochem J ; 473(17): 2573-89, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27574023

ABSTRACT

A major objective in biological research is to understand spatial and temporal requirements for any given gene, especially in dynamic processes acting over short periods, such as catalytically driven reactions, subcellular transport, cell division, cell rearrangement and cell migration. The interrogation of such processes requires the use of rapid and flexible methods of interfering with gene function. However, many of the most widely used interventional approaches, such as RNAi or CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 (CRISPR-associated 9), operate at the level of the gene or its transcripts, meaning that the effects of gene perturbation are exhibited over longer time frames than the process under investigation. There has been much activity over the last few years to address this fundamental problem. In the present review, we describe recent advances in disruption technologies acting at the level of the expressed protein, involving inducible methods of protein cleavage, (in)activation, protein sequestration or degradation. Drawing on examples from model organisms we illustrate the utility of fast-acting techniques and discuss how different components of the molecular toolkit can be employed to dissect previously intractable biochemical processes and cellular behaviours.


Subject(s)
Proteins/physiology , Clustered Regularly Interspaced Short Palindromic Repeats , Humans
19.
Blood ; 121(26): 5218-27, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23704091

ABSTRACT

Missense mutations in transcription factor GATA1 underlie a spectrum of congenital red blood cell and platelet disorders. We investigated how these alterations cause distinct clinical phenotypes by combining structural, biochemical, and genomic approaches with gene complementation systems that examine GATA1 function in biologically relevant cellular contexts. Substitutions that disrupt FOG1 cofactor binding impair both gene activation and repression and are associated with pronounced clinical phenotypes. Moreover, clinical severity correlates with the degree of FOG1 disruption. Surprisingly, 2 mutations shown to impair DNA binding of GATA1 in vitro did not measurably affect in vivo target gene occupancy. Rather, one of these disrupted binding to the TAL1 complex, implicating it in diseases caused by GATA1 mutations. Diminished TAL1 complex recruitment mainly impairs transcriptional activation and is linked to relatively mild disease. Notably, different substitutions at the same amino acid can selectively inhibit TAL1 complex or FOG1 binding, producing distinct cellular and clinical phenotypes. The structure-function relationships elucidated here were not predicted by prior in vitro or computational studies. Thus, our findings uncover novel disease mechanisms underlying GATA1 mutations and highlight the power of gene complementation assays for elucidating the molecular basis of genetic diseases.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , GATA1 Transcription Factor/genetics , Genetic Complementation Test , Hematologic Diseases/etiology , Mutation, Missense/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , Cell Proliferation , Chromatin Immunoprecipitation , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/chemistry , GATA1 Transcription Factor/metabolism , Gene Expression Profiling , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , T-Cell Acute Lymphocytic Leukemia Protein 1 , Transcription Factors/chemistry , Transcription Factors/genetics
20.
Proc Natl Acad Sci U S A ; 108(22): E159-68, 2011 May 31.
Article in English | MEDLINE | ID: mdl-21536911

ABSTRACT

Acetylation of histones triggers association with bromodomain-containing proteins that regulate diverse chromatin-related processes. Although acetylation of transcription factors has been appreciated for some time, the mechanistic consequences are less well understood. The hematopoietic transcription factor GATA1 is acetylated at conserved lysines that are required for its stable association with chromatin. We show that the BET family protein Brd3 binds via its first bromodomain (BD1) to GATA1 in an acetylation-dependent manner in vitro and in vivo. Mutation of a single residue in BD1 that is involved in acetyl-lysine binding abrogated recruitment of Brd3 by GATA1, demonstrating that acetylation of GATA1 is essential for Brd3 association with chromatin. Notably, Brd3 is recruited by GATA1 to both active and repressed target genes in a fashion seemingly independent of histone acetylation. Anti-Brd3 ChIP followed by massively parallel sequencing in GATA1-deficient erythroid precursor cells and those that are GATA1 replete revealed that GATA1 is a major determinant of Brd3 recruitment to genomic targets within chromatin. A pharmacologic compound that occupies the acetyl-lysine binding pockets of Brd3 bromodomains disrupts the Brd3-GATA1 interaction, diminishes the chromatin occupancy of both proteins, and inhibits erythroid maturation. Together these findings provide a mechanism for GATA1 acetylation and suggest that Brd3 "reads" acetyl marks on nuclear factors to promote their stable association with chromatin.


Subject(s)
Erythroid Cells/cytology , GATA1 Transcription Factor/chemistry , Gene Expression Regulation , Histones/chemistry , Nuclear Proteins/chemistry , Acetylation , Animals , Chromatin/chemistry , Chromatin/metabolism , Chromatin Immunoprecipitation , GATA1 Transcription Factor/metabolism , Hematopoiesis , Mice , Mutation , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL