Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
Add more filters

Publication year range
1.
Cell ; 173(2): 430-442.e17, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29606353

ABSTRACT

Fetal hemoglobin (HbF, α2γ2) level is genetically controlled and modifies severity of adult hemoglobin (HbA, α2ß2) disorders, sickle cell disease, and ß-thalassemia. Common genetic variation affects expression of BCL11A, a regulator of HbF silencing. To uncover how BCL11A supports the developmental switch from γ- to ß- globin, we use a functional assay and protein binding microarray to establish a requirement for a zinc-finger cluster in BCL11A in repression and identify a preferred DNA recognition sequence. This motif appears in embryonic and fetal-expressed globin promoters and is duplicated in γ-globin promoters. The more distal of the duplicated motifs is mutated in individuals with hereditary persistence of HbF. Using the CUT&RUN approach to map protein binding sites in erythroid cells, we demonstrate BCL11A occupancy preferentially at the distal motif, which can be disrupted by editing the promoter. Our findings reveal that direct γ-globin gene promoter repression by BCL11A underlies hemoglobin switching.


Subject(s)
Carrier Proteins/metabolism , Fetal Hemoglobin/genetics , Nuclear Proteins/metabolism , Base Sequence , Binding Sites , Carrier Proteins/genetics , Cell Line , Chromatin/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Gene Editing , Humans , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/metabolism , Repressor Proteins , Zinc Fingers/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/pathology , gamma-Globins/genetics
2.
Nat Rev Mol Cell Biol ; 20(4): 199-210, 2019 04.
Article in English | MEDLINE | ID: mdl-30824861

ABSTRACT

The tumour suppressor p53 has a central role in the response to cellular stress. Activated p53 transcriptionally regulates hundreds of genes that are involved in multiple biological processes, including in DNA damage repair, cell cycle arrest, apoptosis and senescence. In the context of DNA damage, p53 is thought to be a decision-making transcription factor that selectively activates genes as part of specific gene expression programmes to determine cellular outcomes. In this Review, we discuss the multiple molecular mechanisms of p53 regulation and how they modulate the induction of apoptosis or cell cycle arrest following DNA damage. Specifically, we discuss how the interaction of p53 with DNA and chromatin affects gene expression, and how p53 post-translational modifications, its temporal expression dynamics and its interactions with chromatin regulators and transcription factors influence cell fate. These multiple layers of regulation enable p53 to execute cellular responses that are appropriate for specific cellular states and environmental conditions.


Subject(s)
Apoptosis/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle Checkpoints/genetics , DNA Damage/genetics , Gene Expression Regulation/genetics , Humans
3.
Mol Cell ; 77(2): 324-337.e8, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31704182

ABSTRACT

A major challenge in biology is to understand how complex gene expression patterns are encoded in the genome. While transcriptional enhancers have been studied extensively, few transcriptional silencers have been identified, and they remain poorly understood. Here, we used a novel strategy to screen hundreds of sequences for tissue-specific silencer activity in whole Drosophila embryos. Almost all of the transcriptional silencers that we identified were also active enhancers in other cellular contexts. These elements are bound by more transcription factors than non-silencers. A subset of these silencers forms long-range contacts with promoters. Deletion of a silencer caused derepression of its target gene. Our results challenge the common practice of treating enhancers and silencers as separate classes of regulatory elements and suggest the possibility that thousands or more bifunctional CRMs remain to be discovered in Drosophila and 104-105 in humans.


Subject(s)
Drosophila/genetics , Enhancer Elements, Genetic/genetics , Silencer Elements, Transcriptional/genetics , Transcription, Genetic/genetics , Animals , Animals, Genetically Modified/genetics , Male
4.
Mol Cell ; 74(2): 245-253.e6, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30826165

ABSTRACT

Transcription factors (TFs) control gene expression by binding DNA recognition sites in genomic regulatory regions. Although most forkhead TFs recognize a canonical forkhead (FKH) motif, RYAAAYA, some forkheads recognize a completely different (FHL) motif, GACGC. Bispecific forkhead proteins recognize both motifs, but the molecular basis for bispecific DNA recognition is not understood. We present co-crystal structures of the FoxN3 DNA binding domain bound to the FKH and FHL sites, respectively. FoxN3 adopts a similar conformation to recognize both motifs, making contacts with different DNA bases using the same amino acids. However, the DNA structure is different in the two complexes. These structures reveal how a single TF binds two unrelated DNA sequences and the importance of DNA shape in the mechanism of bispecific recognition.


Subject(s)
Cell Cycle Proteins/chemistry , DNA-Binding Proteins/chemistry , DNA/chemistry , Nucleic Acid Conformation , Repressor Proteins/chemistry , Amino Acid Sequence/genetics , Base Sequence/genetics , Binding Sites/genetics , Cell Cycle Proteins/genetics , Crystallography, X-Ray , DNA/genetics , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Gene Expression Regulation/genetics , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nucleotide Motifs/genetics , Regulatory Sequences, Nucleic Acid/genetics , Repressor Proteins/genetics
5.
Genes Dev ; 32(9-10): 723-736, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29764918

ABSTRACT

The K50 (lysine at amino acid position 50) homeodomain (HD) protein Orthodenticle (Otd) is critical for anterior patterning and brain and eye development in most metazoans. In Drosophila melanogaster, another K50HD protein, Bicoid (Bcd), has evolved to replace Otd's ancestral function in embryo patterning. Bcd is distributed as a long-range maternal gradient and activates transcription of a large number of target genes, including otd Otd and Bcd bind similar DNA sequences in vitro, but how their transcriptional activities are integrated to pattern anterior regions of the embryo is unknown. Here we define three major classes of enhancers that are differentially sensitive to binding and transcriptional activation by Bcd and Otd. Class 1 enhancers are initially activated by Bcd, and activation is transferred to Otd via a feed-forward relay (FFR) that involves sequential binding of the two proteins to the same DNA motif. Class 2 enhancers are activated by Bcd and maintained by an Otd-independent mechanism. Class 3 enhancers are never bound by Bcd, but Otd binds and activates them in a second wave of zygotic transcription. The specific activities of enhancers in each class are mediated by DNA motif variants preferentially bound by Bcd or Otd and the presence or absence of sites for cofactors that interact with these proteins. Our results define specific patterning roles for Bcd and Otd and provide mechanisms for coordinating the precise timing of gene expression patterns during embryonic development.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Amino Acid Motifs , Animals , Body Patterning/genetics , Drosophila melanogaster/metabolism , Embryonic Development/drug effects , Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Protein Binding
6.
Nucleic Acids Res ; 51(2): 574-594, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36537216

ABSTRACT

The lysine acetyltransferase KAT6A (MOZ, MYST3) belongs to the MYST family of chromatin regulators, facilitating histone acetylation. Dysregulation of KAT6A has been implicated in developmental syndromes and the onset of acute myeloid leukemia (AML). Previous work suggests that KAT6A is recruited to its genomic targets by a combinatorial function of histone binding PHD fingers, transcription factors and chromatin binding interaction partners. Here, we demonstrate that a winged helix (WH) domain at the very N-terminus of KAT6A specifically interacts with unmethylated CpG motifs. This DNA binding function leads to the association of KAT6A with unmethylated CpG islands (CGIs) genome-wide. Mutation of the essential amino acids for DNA binding completely abrogates the enrichment of KAT6A at CGIs. In contrast, deletion of a second WH domain or the histone tail binding PHD fingers only subtly influences the binding of KAT6A to CGIs. Overexpression of a KAT6A WH1 mutant has a dominant negative effect on H3K9 histone acetylation, which is comparable to the effects upon overexpression of a KAT6A HAT domain mutant. Taken together, our work revealed a previously unrecognized chromatin recruitment mechanism of KAT6A, offering a new perspective on the role of KAT6A in gene regulation and human diseases.


Subject(s)
Chromatin , Histone Acetyltransferases , Histones , Humans , Chromatin/genetics , CpG Islands/genetics , DNA , Histone Acetyltransferases/metabolism , Histones/metabolism , Acetylation
7.
Trends Genet ; 37(6): 514-527, 2021 06.
Article in English | MEDLINE | ID: mdl-33712326

ABSTRACT

Silencers are regulatory DNA elements that reduce transcription from their target promoters; they are the repressive counterparts of enhancers. Although discovered decades ago, and despite evidence of their importance in development and disease, silencers have been much less studied than enhancers. Recently, however, a series of papers have reported systematic studies of silencers in various model systems. Silencers are often bifunctional regulatory elements that can also act as enhancers, depending on cellular context, and are enriched for expression quantitative trait loci (eQTLs) and disease-associated variants. There is not yet evidence of a 'silencer chromatin signature', in the distribution of histone modifications or associated proteins, that is common to all silencers; instead, silencers may fall into various subclasses, acting by distinct (and possibly overlapping) mechanisms.


Subject(s)
Gene Expression Regulation , Gene Silencing , Regulatory Sequences, Nucleic Acid/genetics , Animals , Chromatin/genetics , Evolution, Molecular , Genetic Techniques , Humans , Promoter Regions, Genetic
8.
Genome Res ; 31(9): 1663-1679, 2021 09.
Article in English | MEDLINE | ID: mdl-34426512

ABSTRACT

Antibodies offer a powerful means to interrogate specific proteins in a complex milieu. However, antibody availability and reliability can be problematic, whereas epitope tagging can be impractical in many cases. To address these limitations, the Protein Capture Reagents Program (PCRP) generated over a thousand renewable monoclonal antibodies (mAbs) against human presumptive chromatin proteins. However, these reagents have not been widely field-tested. We therefore performed a screen to test their ability to enrich genomic regions via chromatin immunoprecipitation (ChIP) and a variety of orthogonal assays. Eight hundred eighty-seven unique antibodies against 681 unique human transcription factors (TFs) were assayed by ultra-high-resolution ChIP-exo/seq, generating approximately 1200 ChIP-exo data sets, primarily in a single pass in one cell type (K562). Subsets of PCRP mAbs were further tested in ChIP-seq, CUT&RUN, STORM super-resolution microscopy, immunoblots, and protein binding microarray (PBM) experiments. About 5% of the tested antibodies displayed high-confidence target (i.e., cognate antigen) enrichment across at least one assay and are strong candidates for additional validation. An additional 34% produced ChIP-exo data that were distinct from background and thus warrant further testing. The remaining 61% were not substantially different from background, and likely require consideration of a much broader survey of cell types and/or assay optimizations. We show and discuss the metrics and challenges to antibody validation in chromatin-based assays.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Transcription Factors , Binding Sites , Chromatin Immunoprecipitation , Humans , Indicators and Reagents , Reproducibility of Results , Transcription Factors/metabolism
9.
Development ; 148(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34423346

ABSTRACT

During convergent differentiation, multiple developmental lineages produce a highly similar or identical cell type. However, few molecular players that drive convergent differentiation are known. Here, we show that the C. elegans Forkhead transcription factor UNC-130 is required in only one of three convergent lineages that produce the same glial cell type. UNC-130 acts transiently as a repressor in progenitors and newly-born terminal cells to allow the proper specification of cells related by lineage rather than by cell type or function. Specification defects correlate with UNC-130:DNA binding, and UNC-130 can be functionally replaced by its human homolog, the neural crest lineage determinant FoxD3. We propose that, in contrast to terminal selectors that activate cell type-specific transcriptional programs in terminally differentiating cells, UNC-130 acts early and specifically in one convergent lineage to produce a cell type that also arises from molecularly distinct progenitors in other lineages.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Lineage , Neuroglia/metabolism , Transcription Factors/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cell Differentiation , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Neuroglia/cytology , Transcription Factors/genetics
11.
Cell ; 138(2): 314-27, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19632181

ABSTRACT

Differences in expression, protein interactions, and DNA binding of paralogous transcription factors ("TF parameters") are thought to be important determinants of regulatory and biological specificity. However, both the extent of TF divergence and the relative contribution of individual TF parameters remain undetermined. We comprehensively identify dimerization partners, spatiotemporal expression patterns, and DNA-binding specificities for the C. elegans bHLH family of TFs, and model these data into an integrated network. This network displays both specificity and promiscuity, as some bHLH proteins, DNA sequences, and tissues are highly connected, whereas others are not. By comparing all bHLH TFs, we find extensive divergence and that all three parameters contribute equally to bHLH divergence. Our approach provides a framework for examining divergence for other protein families in C. elegans and in other complex multicellular organisms, including humans. Cross-species comparisons of integrated networks may provide further insights into molecular features underlying protein family evolution. For a video summary of this article, see the PaperFlick file available with the online Supplemental Data.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , DNA/metabolism , Gene Regulatory Networks , Male , Molecular Sequence Data , Promoter Regions, Genetic , Protein Multimerization
12.
Genome Res ; 30(5): 736-748, 2020 05.
Article in English | MEDLINE | ID: mdl-32424069

ABSTRACT

Deciphering the interplay between chromatin accessibility and transcription factor (TF) binding is fundamental to understanding transcriptional regulation, control of cellular states, and the establishment of new phenotypes. Recent genome-wide chromatin accessibility profiling studies have provided catalogs of putative open regions, where TFs can recognize their motifs and regulate gene expression programs. Here, we present motif enrichment in differential elements of accessibility (MEDEA), a computational tool that analyzes high-throughput chromatin accessibility genomic data to identify cell-type-specific accessible regions and lineage-specific motifs associated with TF binding therein. To benchmark MEDEA, we used a panel of reference cell lines profiled by ENCODE and curated by the ENCODE Project Consortium for the ENCODE-DREAM Challenge. By comparing results with RNA-seq data, ChIP-seq peaks, and DNase-seq footprints, we show that MEDEA improves the detection of motifs associated with known lineage specifiers. We then applied MEDEA to 610 ENCODE DNase-seq data sets, where it revealed significant motifs even when absolute enrichment was low and where it identified novel regulators, such as NRF1 in kidney development. Finally, we show that MEDEA performs well on both bulk and single-cell ATAC-seq data. MEDEA is publicly available as part of our Glossary-GENRE suite for motif enrichment analysis.


Subject(s)
Chromatin/metabolism , Regulatory Elements, Transcriptional , Sequence Analysis, DNA/methods , Transcription Factors/metabolism , Binding Sites , Cell Line , Cell Lineage/genetics , DNA/chemistry , Humans , Nucleotide Motifs
13.
Mol Syst Biol ; 18(8): e10473, 2022 08.
Article in English | MEDLINE | ID: mdl-35996956

ABSTRACT

Neuronal stimulation induced by the brain-derived neurotrophic factor (BDNF) triggers gene expression, which is crucial for neuronal survival, differentiation, synaptic plasticity, memory formation, and neurocognitive health. However, its role in chromatin regulation is unclear. Here, using temporal profiling of chromatin accessibility and transcription in mouse primary cortical neurons upon either BDNF stimulation or depolarization (KCl), we identify features that define BDNF-specific chromatin-to-gene expression programs. Enhancer activation is an early event in the regulatory control of BDNF-treated neurons, where the bZIP motif-binding Fos protein pioneered chromatin opening and cooperated with co-regulatory transcription factors (Homeobox, EGRs, and CTCF) to induce transcription. Deleting cis-regulatory sequences affect BDNF-mediated Arc expression, a regulator of synaptic plasticity. BDNF-induced accessible regions are linked to preferential exon usage by neurodevelopmental disorder-related genes and the heritability of neuronal complex traits, which were validated in human iPSC-derived neurons. Thus, we provide a comprehensive view of BDNF-mediated genome regulatory features using comparative genomic approaches to dissect mammalian neuronal stimulation.


Subject(s)
Brain-Derived Neurotrophic Factor , Chromatin , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Chromatin/genetics , Chromatin/metabolism , Humans , Mammals/genetics , Mice , Neurons/metabolism , Transcription Factors/metabolism
14.
Nat Immunol ; 13(1): 95-102, 2011 Nov 20.
Article in English | MEDLINE | ID: mdl-22101729

ABSTRACT

The unique DNA-binding properties of distinct NF-κB dimers influence the selective regulation of NF-κB target genes. To more thoroughly investigate these dimer-specific differences, we combined protein-binding microarrays and surface plasmon resonance to evaluate DNA sites recognized by eight different NF-κB dimers. We observed three distinct binding-specificity classes and clarified mechanisms by which dimers might regulate distinct sets of genes. We identified many new nontraditional NF-κB binding site (κB site) sequences and highlight the plasticity of NF-κB dimers in recognizing κB sites with a single consensus half-site. This study provides a database that can be used in efforts to identify NF-κB target sites and uncover gene regulatory circuitry.


Subject(s)
Gene Expression Regulation , NF-kappa B/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cluster Analysis , DNA/chemistry , DNA/genetics , Databases, Genetic , Humans , Macrophages/metabolism , Mice , NF-kappa B/genetics , Protein Array Analysis , Protein Binding , Protein Multimerization
15.
Cell ; 133(7): 1266-76, 2008 Jun 27.
Article in English | MEDLINE | ID: mdl-18585359

ABSTRACT

Most homeodomains are unique within a genome, yet many are highly conserved across vast evolutionary distances, implying strong selection on their precise DNA-binding specificities. We determined the binding preferences of the majority (168) of mouse homeodomains to all possible 8-base sequences, revealing rich and complex patterns of sequence specificity and showing that there are at least 65 distinct homeodomain DNA-binding activities. We developed a computational system that successfully predicts binding sites for homeodomain proteins as distant from mouse as Drosophila and C. elegans, and we infer full 8-mer binding profiles for the majority of known animal homeodomains. Our results provide an unprecedented level of resolution in the analysis of this simple domain structure and suggest that variation in sequence recognition may be a factor in its functional diversity and evolutionary success.


Subject(s)
DNA/chemistry , Homeodomain Proteins/chemistry , Animals , Base Sequence , Computational Biology , Conserved Sequence , DNA/metabolism , Evolution, Molecular , Homeodomain Proteins/metabolism , Mice , Models, Molecular , Protein Binding , Transcription Factors/chemistry , Transcription Factors/metabolism
16.
Nature ; 549(7671): 287-291, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28869966

ABSTRACT

The Polycomb repressive complex 2 (PRC2) mainly mediates transcriptional repression and has essential roles in various biological processes including the maintenance of cell identity and proper differentiation. Polycomb-like (PCL) proteins, such as PHF1, MTF2 and PHF19, are PRC2-associated factors that form sub-complexes with PRC2 core components, and have been proposed to modulate the enzymatic activity of PRC2 or the recruitment of PRC2 to specific genomic loci. Mammalian PRC2-binding sites are enriched in CG content, which correlates with CpG islands that display a low level of DNA methylation. However, the mechanism of PRC2 recruitment to CpG islands is not fully understood. Here we solve the crystal structures of the N-terminal domains of PHF1 and MTF2 with bound CpG-containing DNAs in the presence of H3K36me3-containing histone peptides. We show that the extended homologous regions of both proteins fold into a winged-helix structure, which specifically binds to the unmethylated CpG motif but in a completely different manner from the canonical winged-helix DNA recognition motif. We also show that the PCL extended homologous domains are required for efficient recruitment of PRC2 to CpG island-containing promoters in mouse embryonic stem cells. Our research provides the first, to our knowledge, direct evidence to demonstrate that PCL proteins are crucial for PRC2 recruitment to CpG islands, and further clarifies the roles of these proteins in transcriptional regulation in vivo.


Subject(s)
CpG Islands/genetics , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Animals , Binding Sites , Chromatin/chemistry , Chromatin/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Histones/chemistry , Histones/metabolism , Humans , Mice , Models, Molecular , Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Domains , Transcription Factors/chemistry , Transcription Factors/metabolism , Transcription, Genetic
17.
Proc Natl Acad Sci U S A ; 117(28): 16516-16526, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601179

ABSTRACT

LIN28B is highly expressed in neuroblastoma and promotes tumorigenesis, at least, in part, through inhibition of let-7 microRNA biogenesis. Here, we report that overexpression of either wild-type (WT) LIN28B or a LIN28B mutant that is unable to inhibit let-7 processing increases the penetrance of MYCN-induced neuroblastoma, potentiates the invasion and migration of transformed sympathetic neuroblasts, and drives distant metastases in vivo. Genome-wide chromatin immunoprecipitation coupled with massively parallel DNA sequencing (ChIP-seq) and coimmunoprecipitation experiments show that LIN28B binds active gene promoters in neuroblastoma cells through protein-protein interaction with the sequence-specific zinc-finger transcription factor ZNF143 and activates the expression of downstream targets, including transcription factors forming the adrenergic core regulatory circuitry that controls the malignant cell state in neuroblastoma as well as GSK3B and L1CAM that are involved in neuronal cell adhesion and migration. These findings reveal an unexpected let-7-independent function of LIN28B in transcriptional regulation during neuroblastoma pathogenesis.


Subject(s)
N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , RNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , Animals, Genetically Modified , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/physiopathology , Protein Binding , RNA-Binding Proteins/genetics , Trans-Activators/genetics , Zebrafish
18.
Mol Cell ; 55(4): 640-8, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25042805

ABSTRACT

A major challenge in obtaining a full molecular description of evolutionary adaptation is to characterize how transcription factor (TF) DNA-binding specificity can change. To identify mechanisms of TF diversification, we performed detailed comparisons of yeast C2H2 ZF proteins with identical canonical recognition residues that are expected to bind the same DNA sequences. Unexpectedly, we found that ZF proteins can adapt to recognize new binding sites in a modular fashion whereby binding to common core sites remains unaffected. We identified two distinct mechanisms, conserved across multiple Ascomycota species, by which this molecular adaptation occurred. Our results suggest a route for TF evolution that alleviates negative pleiotropic effects by modularly gaining new binding sites. These findings expand our current understanding of ZF DNA binding and provide evidence for paralogous ZFs utilizing alternate modes of DNA binding to recognize unique sets of noncanonical binding sites.


Subject(s)
Ascomycota/metabolism , DNA, Fungal/genetics , DNA-Binding Proteins/metabolism , Fungal Proteins/genetics , Transcription Factors/metabolism , Zinc Fingers/physiology , Amino Acid Motifs , Ascomycota/classification , Ascomycota/genetics , Conserved Sequence , DNA-Binding Proteins/chemistry , Evolution, Molecular , Fungal Proteins/metabolism , Genome, Fungal , Zinc Fingers/genetics
19.
Proc Natl Acad Sci U S A ; 116(29): 14614-14619, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31262815

ABSTRACT

Aberrant MYC oncogene activation is one of the most prevalent characteristics of cancer. By overlapping datasets of Drosophila genes that are insulin-responsive and also regulate nucleolus size, we enriched for Myc target genes required for cellular biosynthesis. Among these, we identified the aminoacyl tRNA synthetases (aaRSs) as essential mediators of Myc growth control in Drosophila and found that their pharmacologic inhibition is sufficient to kill MYC-overexpressing human cells, indicating that aaRS inhibitors might be used to selectively target MYC-driven cancers. We suggest a general principle in which oncogenic increases in cellular biosynthesis sensitize cells to disruption of protein homeostasis.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/drug therapy , Transcription Factors/metabolism , Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Animals , Animals, Genetically Modified , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Epithelial Cells , Female , Humans , Insulin/metabolism , Male , Neoplasms/genetics , Neoplasms/pathology , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/genetics
20.
Genes Dev ; 27(14): 1551-6, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23873939

ABSTRACT

The Drosophila male-specific lethal (MSL) dosage compensation complex increases transcript levels on the single male X chromosome to equal the transcript levels in XX females. However, it is not known how the MSL complex is linked to its DNA recognition elements, the critical first step in dosage compensation. Here, we demonstrate that a previously uncharacterized zinc finger protein, CLAMP (chromatin-linked adaptor for MSL proteins), functions as the first link between the MSL complex and the X chromosome. CLAMP directly binds to the MSL complex DNA recognition elements and is required for the recruitment of the MSL complex. The discovery of CLAMP identifies a key factor required for the chromosome-specific targeting of dosage compensation, providing new insights into how subnuclear domains of coordinate gene regulation are formed within metazoan genomes.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dosage Compensation, Genetic , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , X Chromosome/genetics , X Chromosome/metabolism , Animals , Cell Line , Female , Male , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL