Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37090641

ABSTRACT

Reprogramming of the gamete into a developmentally competent embryo identity is a fundamental aspect of preimplantation development. One of the most important processes of this reprogramming is the transcriptional awakening during embryonic genome activation (EGA), which robustly occurs in fertilized embryos but is defective in most somatic cell nuclear transfer (SCNT) embryos. However, little is known about the genome-wide underlying chromatin landscape during EGA in SCNT embryos and how it differs from a fertilized embryo. By profiling open chromatin genome-wide in both types of bovine embryos, we find that SCNT embryos fail to reprogram a subset of the EGA gene targets that are normally activated in fertilized embryos. Importantly, a small number of transcription factor (TF) motifs explain most chromatin regions that fail to open in SCNT embryos suggesting that over-expression of a limited number of TFs may provide more robust reprogramming. One such TF, the zygotically-expressed bovine gene DUXC which is a homologue of EGA factors DUX/DUX4 in mouse/human, is alone capable of activating ∻84% of all EGA transcripts that fail to activate normally in SCNT embryos. Additionally, single-cell chromatin profiling revealed low intra-embryo heterogeneity but high inter-embryo heterogeneity in SCNT embryos and an uncoupling of cell division and open chromatin reprogramming during EGA. Surprisingly, our data also indicate that transcriptional defects may arise downstream of promoter chromatin opening in SCNT embryos, suggesting additional mechanistic insights into how and why transcription at EGA is dysregulated. We anticipate that our work will lead to altered SCNT protocols to increase the developmental competency of bovine SCNT embryos.

2.
bioRxiv ; 2023 Jun 08.
Article in English | MEDLINE | ID: mdl-37034731

ABSTRACT

The embryonic transcription factor DUX regulates chromatin opening and gene expression in totipotent cleavage-stage mouse embryos, and its expression in embryonic stem cells promotes their conversion to 2-cell embryo-like cells (2CLCs) with extraembryonic potential. However, little is known regarding which domains within mouse DUX interact with particular chromatin and transcription regulators. Here, we reveal that the C-terminus of mouse DUX contains five uncharacterized ~100 amino acid (aa) repeats followed by an acidic 14 amino acid tail. Unexpectedly, structure-function approaches classify two repeats as 'active' and three as 'inactive' in cleavage/2CLC transcription program enhancement, with differences narrowed to a key 6 amino acid section. Our proximity dependent biotin ligation (BioID) approach identified factors selectively associated with active DUX repeat derivatives (including the 14aa 'tail'), including transcription and chromatin factors such as SWI/SNF (BAF) complex, as well as nucleolar factors that have been previously implicated in regulating the Dux locus. Finally, our mechanistic studies reveal cooperativity between DUX active repeats and the acidic tail in cofactor recruitment, DUX target opening, and transcription. Taken together, we provide several new insights into DUX structure-function, and mechanisms of chromatin and gene regulation.

3.
Nat Genet ; 53(8): 1207-1220, 2021 08.
Article in English | MEDLINE | ID: mdl-34267371

ABSTRACT

In mammalian embryos, proper zygotic genome activation (ZGA) underlies totipotent development. Double homeobox (DUX)-family factors participate in ZGA, and mouse Dux is required for forming cultured two-cell (2C)-like cells. Remarkably, in mouse embryonic stem cells, Dux is activated by the tumor suppressor p53, and Dux expression promotes differentiation into expanded-fate cell types. Long-read sequencing and assembly of the mouse Dux locus reveals its complex chromatin regulation including putative positive and negative feedback loops. We show that the p53-DUX/DUX4 regulatory axis is conserved in humans. Furthermore, we demonstrate that cells derived from patients with facioscapulohumeral muscular dystrophy (FSHD) activate human DUX4 during p53 signaling via a p53-binding site in a primate-specific subtelomeric long terminal repeat (LTR)10C element. In summary, our work shows that p53 activation convergently evolved to couple p53 to Dux/DUX4 activation in embryonic stem cells, embryos and cells from patients with FSHD, potentially uniting the developmental and disease regulation of DUX-family factors and identifying evidence-based therapeutic opportunities for FSHD.


Subject(s)
Homeodomain Proteins/genetics , Mouse Embryonic Stem Cells/physiology , Muscular Dystrophy, Facioscapulohumeral/pathology , Tumor Suppressor Protein p53/genetics , Animals , Cell Differentiation/genetics , Cellular Reprogramming , DNA Damage , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Knockout , Mouse Embryonic Stem Cells/cytology , Muscular Dystrophy, Facioscapulohumeral/genetics , Nuclear Proteins/genetics , Pluripotent Stem Cells/physiology , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Zygote/cytology
4.
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
5.
Dev Cell ; 50(5): 658-671.e7, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31327741

ABSTRACT

Advances in cancer immunotherapies make it critical to identify genes that modulate antigen presentation and tumor-immune interactions. We report that DUX4, an early embryonic transcription factor that is normally silenced in somatic tissues, is re-expressed in diverse solid cancers. Both cis-acting inherited genetic variation and somatically acquired mutations in trans-acting repressors contribute to DUX4 re-expression in cancer. Although many DUX4 target genes encode self-antigens, DUX4-expressing cancers were paradoxically characterized by reduced markers of anti-tumor cytolytic activity and lower major histocompatibility complex (MHC) class I gene expression. We demonstrate that DUX4 expression blocks interferon-γ-mediated induction of MHC class I, implicating suppressed antigen presentation in DUX4-mediated immune evasion. Clinical data in metastatic melanoma confirmed that DUX4 expression was associated with significantly reduced progression-free and overall survival in response to anti-CTLA-4. Our results demonstrate that cancers can escape immune surveillance by reactivating a normal developmental pathway and identify a therapeutically relevant mechanism of cell-intrinsic immune evasion.


Subject(s)
Genes, MHC Class I , Homeodomain Proteins/metabolism , Immune Evasion , Neoplasms/immunology , Antigen Presentation , CTLA-4 Antigen/immunology , HeLa Cells , Homeodomain Proteins/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , MCF-7 Cells , Neoplasms/genetics , T-Lymphocytes/immunology
6.
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
7.
Elife ; 72018 03 13.
Article in English | MEDLINE | ID: mdl-29533181

ABSTRACT

The DUX4 transcription factor is encoded by a retrogene embedded in each unit of the D4Z4 macrosatellite repeat. DUX4 is normally expressed in the cleavage-stage embryo, whereas chromatin repression prevents DUX4 expression in most somatic tissues. Failure of this repression causes facioscapulohumeral muscular dystrophy (FSHD) due to mis-expression of DUX4 in skeletal muscle. In this study, we used CRISPR/Cas9 engineered chromatin immunoprecipitation (enChIP) locus-specific proteomics to characterize D4Z4-associated proteins. These and other approaches identified the Nucleosome Remodeling Deacetylase (NuRD) and Chromatin Assembly Factor 1 (CAF-1) complexes as necessary for DUX4 repression in human skeletal muscle cells and induced pluripotent stem (iPS) cells. Furthermore, DUX4-induced expression of MBD3L proteins partly relieved this repression in FSHD muscle cells. Together, these findings identify NuRD and CAF-1 as mediators of DUX4 chromatin repression and suggest a mechanism for the amplification of DUX4 expression in FSHD muscle cells.


Subject(s)
Epigenesis, Genetic , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Chromatin/genetics , Chromatin Assembly Factor-1/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Regulation, Developmental , Gene Silencing , Homeodomain Proteins/chemistry , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/chemistry , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Muscle Fibers, Skeletal/chemistry , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/physiopathology , Nucleosomes/chemistry , Nucleosomes/genetics
8.
Skelet Muscle ; 7(1): 16, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28870238

ABSTRACT

BACKGROUND: Facioscapulohumeral dystrophy (FSHD) is a progressive muscle disease caused by mutations that lead to epigenetic derepression and inappropriate transcription of the double homeobox 4 (DUX4) gene in skeletal muscle. Drugs that enhance the repression of DUX4 and prevent its expression in skeletal muscle cells therefore represent candidate therapies for FSHD. METHODS: We screened an aggregated chemical library enriched for compounds with epigenetic activities and the Pharmakon 1600 library composed of compounds that have reached clinical testing to identify molecules that decrease DUX4 expression as monitored by the levels of DUX4 target genes in FSHD patient-derived skeletal muscle cell cultures. RESULTS: Our screens identified several classes of molecules that include inhibitors of the bromodomain and extra-terminal (BET) family of proteins and agonists of the beta-2 adrenergic receptor. Further studies showed that compounds from these two classes suppress the expression of DUX4 messenger RNA (mRNA) by blocking the activity of bromodomain-containing protein 4 (BRD4) or by increasing cyclic adenosine monophosphate (cAMP) levels, respectively. CONCLUSIONS: These data uncover pathways involved in the regulation of DUX4 expression in somatic cells, provide potential candidate classes of compounds for FSHD therapeutic development, and create an important opportunity for mechanistic studies that may uncover additional therapeutic targets.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Homeodomain Proteins/metabolism , Muscular Dystrophy, Facioscapulohumeral/metabolism , Nuclear Proteins/metabolism , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , Cell Cycle Proteins , Cells, Cultured , Cyclic AMP/metabolism , High-Throughput Screening Assays , Homeodomain Proteins/genetics , Humans , Myoblasts/drug effects , Myoblasts/metabolism
9.
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
11.
Hum Mol Genet ; 25(20): 4419-4431, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28171552

ABSTRACT

Facioscapulohumeral dystrophy (FSHD) is caused by the mis-expression of the double-homeodomain transcription factor DUX4 in skeletal muscle cells. Many different cell culture models have been developed to study the pathophysiology of FSHD, frequently based on endogenous expression of DUX4 in FSHD cells or by mis-expression of DUX4 in control human muscle cells. Although results generated using each model are generally consistent, differences have also been reported, making it unclear which model(s) faithfully recapitulate DUX4 and FSHD biology. In this study, we systematically compared RNA-seq data generated from three different models of FSHD­lentiviral-based DUX4 expression in myoblasts, doxycycline-inducible DUX4 in myoblasts, and differentiated human FSHD myocytes expressing endogenous DUX4­and show that the DUX4-associated gene expression signatures of each dataset are highly correlated (Pearson's correlation coefficient, r ∼ 0.75-0.85). The few robust differences were attributable to different states of cell differentiation and other differences in experimental design. Our study describes a model system for inducible DUX4 expression that enables reproducible and synchronized experiments and validates the fidelity and FSHD relevance of multiple distinct models of DUX4 expression.


Subject(s)
Homeodomain Proteins/genetics , Models, Biological , Muscular Dystrophy, Facioscapulohumeral/metabolism , Mutation , Myoblasts/metabolism , Transcriptome , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Humans , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/physiopathology , Sequence Analysis, RNA
12.
Epigenetics ; 10(12): 1133-42, 2015.
Article in English | MEDLINE | ID: mdl-26575099

ABSTRACT

Facioscapulohumeral muscular dystrophy is caused by incomplete epigenetic repression of the transcription factor DUX4 in skeletal muscle. A copy of DUX4 is located within each unit of the D4Z4 macrosatellite repeat array and its derepression in somatic cells is caused by either repeat array contraction (FSHD1) or by mutations in the chromatin repressor SMCHD1 (FSHD2). While DUX4 expression has thus far only been detected in FSHD muscle and muscle cell cultures, and increases with in vitro myogenic differentiation, the D4Z4 chromatin structure has only been studied in proliferating myoblasts or non-myogenic cells. We here show that SMCHD1 protein levels at D4Z4 decline during muscle cell differentiation and correlate with DUX4 derepression. In FSHD2, but not FSHD1, the loss of SMCHD1 repressor activity is partially compensated by increased Polycomb Repressive Complex 2 (PRC2)-mediated H3K27 trimethylation at D4Z4, a situation that can be mimicked by SMCHD1 knockdown in control myotubes. In contrast, moderate overexpression of SMCHD1 results in DUX4 silencing in FSHD1 and FSHD2 myotubes demonstrating that DUX4 derepression in FSHD is reversible. Together, we show that in FSHD1 and FSHD2 the decline in SMCHD1 protein levels during muscle cell differentiation renders skeletal muscle sensitive to DUX4.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Muscle Development/genetics , Muscular Dystrophy, Facioscapulohumeral/genetics , Cell Differentiation/genetics , Cell Line , Chromosomal Proteins, Non-Histone/genetics , DNA Methylation , Gene Expression Regulation , Histone Code , Homeodomain Proteins/genetics , Humans , Muscle, Skeletal/metabolism
13.
EMBO J ; 30(8): 1473-84, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21448134

ABSTRACT

Although regulation of histone methylation is believed to contribute to embryonic stem cell (ESC) self-renewal, the mechanisms remain obscure. We show here that the histone H3 trimethyl lysine 4 (H3K4me3) demethylase, KDM5B, is a downstream Nanog target and critical for ESC self-renewal. Although KDM5B is believed to function as a promoter-bound repressor, we find that it paradoxically functions as an activator of a gene network associated with self-renewal. ChIP-Seq reveals that KDM5B is predominantly targeted to intragenic regions and that it is recruited to H3K36me3 via an interaction with the chromodomain protein MRG15. Depletion of KDM5B or MRG15 increases intragenic H3K4me3, increases cryptic intragenic transcription, and inhibits transcriptional elongation of KDM5B target genes. We propose that KDM5B activates self-renewal-associated gene expression by repressing cryptic initiation and maintaining an H3K4me3 gradient important for productive transcriptional elongation.


Subject(s)
DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Transcription, Genetic , Animals , Biomarkers/metabolism , Blotting, Western , Cell Cycle , Cell Proliferation , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , Gene Expression Profiling , Gene Expression Regulation , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Lysine/metabolism , Mice , Nanog Homeobox Protein , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...