DUX4

14,000,000 Leading Edge Experts on the ideXlab platform

Scan Science and Technology

Contact Leading Edge Experts & Companies

Scan Science and Technology

Contact Leading Edge Experts & Companies

The Experts below are selected from a list of 5076 Experts worldwide ranked by ideXlab platform

Rabi Tawil - One of the best experts on this subject based on the ideXlab platform.

  • relationship of DUX4 and target gene expression in fshd myocytes
    Human Mutation, 2021
    Co-Authors: Jonathan Chau, Rabi Tawil, Xiangduo Kong, Nam Nguyen, Katherine Williams, Miya Ball, Tohru Kiyono, Ali Mortazavi, Kyoko Yokomori
    Abstract:

    Facioscapulohumeral dystrophy (FSHD) is associated with the upregulation of the DUX4 transcription factor and its target genes. However, low-frequency DUX4 upregulation in patient myocytes is difficult to detect and examining the relationship and dynamics of DUX4 and target gene expression has been challenging. Using RNAScope in situ hybridization with highly specific probes, we detect the endogenous DUX4 and target gene transcripts in situ in patient skeletal myotubes during 13-day differentiation in vitro. We found that the endogenous DUX4 transcripts primarily localize as foci in one or two nuclei as compared with the accumulation of the recombinant DUX4 transcripts in the cytoplasm. We also found the continuous increase of DUX4 and target gene-positive myotubes after Day 3, arguing against its expected immediate cytotoxicity. Interestingly, DUX4 and target gene expression become discordant later in differentiation with the increase of DUX4-positive/target gene-negative as well as DUX4-negative/target gene-positive myotubes. Depletion of DUX4-activated transcription factors, DUXA and LEUTX, specifically repressed a DUX4-target gene, KDM4E, later in differentiation, suggesting that after the initial activation by DUX4, target genes themselves contribute to the maintenance of downstream gene expression. Together, the study provides important new insights into the dynamics of the DUX4 transcriptional network in FSHD patient myocytes.

  • relationship of DUX4 and target gene expression in fshd myocytes
    bioRxiv, 2020
    Co-Authors: Jonathan Chau, Rabi Tawil, Nam Nguyen, Katherine Williams, Tohru Kiyono, Ali Mortazavi, Xiangduk Kong, Kyoko Yokomori
    Abstract:

    Facioscapulohumeral dystrophy (FSHD) is linked to misexpression of the transcription factor, DUX4. Although DUX4 target gene expression is often readily detectable, analysis of DUX4 expression has been limited due to its low expression in patient samples. Recently, single cell/nucleus RNA-sequencing was used to detect the native expression of DUX4 for the first time, but important spatial relationships with its target gene expression was missing. Furthermore, dynamics of DUX4 expression during myoblast differentiation has not been fully explored. In order to study the spatiotemporal relationship of DUX4 and key target genes, we performed RNA FISH on immortalized FSHD2 patient skeletal muscle cells. Using two probe sets, DUX4 transcripts were detected in 1-4% of myotubes after 3-day differentiation in vitro. We found that DUX4 transcripts mainly localize as foci in one or two nuclei in a myotube compared to abundant accumulation of the target gene transcripts in the cytoplasm. Over a 13-day differentiation timecourse, DUX4 expression without target gene expression significantly increased and peaked at day 7. Target gene expression correlates better with DUX4 expression early in differentiation while the expression of target genes without detectable DUX4 transcripts increases later. Consistently, shRNA depletion of DUX4-activated transcription factors, DUXA and LEUTX, specifically repressed a DUX4-target gene, KDM4E, later in differentiation, suggesting that following the initial activation by DUX4, target genes themselves contribute to the maintenance of downstream gene expression. Together, in situ detection of the DUX4 and target gene transcripts provided new insight into dynamics of DUX4 transcriptional network in FSHD patient myocytes. Significance StatementFSHD is the third most common muscular dystrophy and is associated with upregulation of DUX4, a transcription factor, and its target genes. Although target genes are easily detectable in FSHD, low frequency DUX4 upregulation in patient myocytes is difficult to detect, and examining the relationship and dynamics of DUX4 and target gene expression without artificial overexpression of DUX4 has been challenging. Using RNAScope with highly specific probes, we detect the endogenous DUX4 and target gene transcripts in situ in patient skeletal myotubes during differentiation in vitro. Our study reveals a unique DUX4 expression pattern and its relationship to the expression of target genes, and evidence for self-sustainability of the target gene network. The study provides important new insights into the FSHD disease mechanism.

  • clinically advanced p38 inhibitors suppress DUX4 expression in cellular and animal models of facioscapulohumeral muscular dystrophy
    Journal of Pharmacology and Experimental Therapeutics, 2019
    Co-Authors: Jonathan Oliva, Stephen J. Tapscott, Rabi Tawil, Amy E Campbell, Jun Wen Zhong, Scott Galasinski, Amelia Richey, Marvin J Meyers, Neal Modi, Francis M Sverdrup
    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 β-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 p38β 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 p38β 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.

  • single cell rna sequencing in facioscapulohumeral muscular dystrophy disease etiology and development
    Human Molecular Genetics, 2019
    Co-Authors: Anita Van Den Heuvel, Stephen J. Tapscott, Rabi Tawil, Ahmed Mahfouz, Susan L Kloet, Judit Balog, Baziel G M Van Engelen, Silvere M Van Der Maarel
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) is characterized by sporadic de-repression of the transcription factor DUX4 in skeletal muscle. DUX4 activates a cascade of muscle disrupting events, eventually leading to muscle atrophy and apoptosis. Yet, how sporadic DUX4 expression leads to the generalized muscle wasting remains unclear. Transcriptome analyses have systematically been challenged by the majority of nuclei being DUX4neg, weakening the DUX4 transcriptome signature. Moreover, DUX4 has been shown to be expressed in a highly dynamic burst-like manner, likely resulting in the detection of the downstream cascade of events long after DUX4 expression itself has faded. Identifying the FSHD transcriptome in individual cells and unraveling the cascade of events leading to FSHD development may therefore provide important insights in the disease process. We employed single-cell RNA sequencing, combined with pseudotime trajectory modeling, to study FSHD disease etiology and cellular progression in human primary myocytes. We identified a small FSHD-specific cell population in all tested patient-derived cultures and detected new genes associated with DUX4 de-repression. We furthermore generated an FSHD cellular progression model, reflecting both the early burst-like DUX4 expression as well as the downstream activation of various FSHD-associated pathways, which allowed us to correlate DUX4 expression signature dynamics with that of regulatory complexes, thereby facilitating the prioritization of epigenetic targets for DUX4 silencing. Single-cell transcriptomics combined with pseudotime modeling thus holds valuable information on FSHD disease etiology and progression that can potentially guide biomarker and target selection for therapy.

  • nurd and caf 1 mediated silencing of the d4z4 array is modulated by DUX4 induced mbd3l proteins
    eLife, 2018
    Co-Authors: Amy E Campbell, Rebecca Resnick, Silvère M. Van Der Maarel, Rabi Tawil, Jongwon Lim, Sean C Shadle, Sujatha Jagannathan, Stephen J. Tapscott
    Abstract:

    The DNA sequences of humans and other mammals contain many repetitive regions. This repetition makes these regions difficult to study with conventional approaches, and so the exact role of repetitive DNA is not fully understood. A particular sequence of repetitive DNA that plays an important role in human health contains a gene called DUX4 in each repeat. DUX4 is normally active in stem cells and in early-stage embryos. This gene is then switched off or ‘silenced’ during later stages of development and in most cells of the body. However, in some individuals the DUX4 gene inappropriately activates in muscle cells. This causes a disease known as facioscapulohumeral muscular dystrophy (FSHD), in which muscle weakness begins in the face and upper body and eventually spreads to other muscles. Currently, there is no cure for FSHD. Proteins that bind to DNA can control the activity of nearby genes. Little is known about which proteins silence DUX4 at the appropriate time and in the right cells, so Campbell et al. set out to identify the proteins that attach to the repetitive DNA sequences containing DUX4. Further investigation showed that several of these proteins play an important role in keeping DUX4 turned off, including two protein complexes called NuRD and CAF-1. These complexes are necessary to silence DUX4 in human muscle cells and stem cells. Campbell et al. also identified a protein that can increase the activity of the DUX4 gene in FSHD muscle cells by overcoming the silencing activity of the NuRD complex. Overall, the results presented by Campbell et al. provide the groundwork for developing new treatments for FSHD. The next step will be to discover ways of enhancing the ability of NuRD and CAF-1 to silence the DUX4 gene.

Stephen J. Tapscott - One of the best experts on this subject based on the ideXlab platform.

  • DUX4-Induced Histone Variants H3.X and H3.Y Mark DUX4 Target Genes for Expression.
    Cell reports, 2019
    Co-Authors: Rebecca Resnick, Chao-jen Wong, Danielle C. Hamm, Sean R. Bennett, Peter J. Skene, Sandra B. Hake, Steven Henikoff, Silvère M. Van Der Maarel, Stephen J. Tapscott
    Abstract:

    The DUX4 transcription factor is briefly expressed in the early cleavage-stage embryo, where it induces an early wave of zygotic gene transcription, whereas its mis-expression in skeletal muscle causes the muscular dystrophy facioscapulohumeral dystrophy (FSHD). Here, we show that DUX4 induces the expression of the histone variants H3.X and H3.Y. We have used a myoblast cell line with doxycycline-inducible DUX4 to show that these histone variants are incorporated throughout the body of DUX4-induced genes. Following a brief pulse of DUX4, these histones contribute to greater perdurance and to enhanced re-activation of DUX4 target gene expression. These findings provide a model for H3.X/Y as a chromatin mechanism that facilitates the expression of DUX4 target genes subsequent to a brief pulse of DUX4 expression.

  • DUX4 induced bidirectional hsatii satellite repeat transcripts form intranuclear double stranded rna foci in human cell models of fshd
    Human Molecular Genetics, 2019
    Co-Authors: Chao-jen Wong, Sean R. Bennett, Silvere M Van Der Maarel, Amy E Campbell, Sean C Shadle, Nancy A Karreman, Brenda L Bass, Stephen J. Tapscott
    Abstract:

    : The DUX4 transcription factor is normally expressed in the cleavage stage embryo and regulates genes involved in embryonic genome activation. Mis-expression 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.

  • DUX4 suppresses mhc class i to promote cancer immune evasion and resistance to checkpoint blockade
    Developmental Cell, 2019
    Co-Authors: Guoliang Chew, Stephen J. Tapscott, Amy E Campbell, Sean C Shadle, Emma J De Neef, Nicholas A Sutliff, Robert K Bradley
    Abstract:

    Summary 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.

  • clinically advanced p38 inhibitors suppress DUX4 expression in cellular and animal models of facioscapulohumeral muscular dystrophy
    Journal of Pharmacology and Experimental Therapeutics, 2019
    Co-Authors: Jonathan Oliva, Stephen J. Tapscott, Rabi Tawil, Amy E Campbell, Jun Wen Zhong, Scott Galasinski, Amelia Richey, Marvin J Meyers, Neal Modi, Francis M Sverdrup
    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 β-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 p38β 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 p38β 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.

  • single cell rna sequencing in facioscapulohumeral muscular dystrophy disease etiology and development
    Human Molecular Genetics, 2019
    Co-Authors: Anita Van Den Heuvel, Stephen J. Tapscott, Rabi Tawil, Ahmed Mahfouz, Susan L Kloet, Judit Balog, Baziel G M Van Engelen, Silvere M Van Der Maarel
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) is characterized by sporadic de-repression of the transcription factor DUX4 in skeletal muscle. DUX4 activates a cascade of muscle disrupting events, eventually leading to muscle atrophy and apoptosis. Yet, how sporadic DUX4 expression leads to the generalized muscle wasting remains unclear. Transcriptome analyses have systematically been challenged by the majority of nuclei being DUX4neg, weakening the DUX4 transcriptome signature. Moreover, DUX4 has been shown to be expressed in a highly dynamic burst-like manner, likely resulting in the detection of the downstream cascade of events long after DUX4 expression itself has faded. Identifying the FSHD transcriptome in individual cells and unraveling the cascade of events leading to FSHD development may therefore provide important insights in the disease process. We employed single-cell RNA sequencing, combined with pseudotime trajectory modeling, to study FSHD disease etiology and cellular progression in human primary myocytes. We identified a small FSHD-specific cell population in all tested patient-derived cultures and detected new genes associated with DUX4 de-repression. We furthermore generated an FSHD cellular progression model, reflecting both the early burst-like DUX4 expression as well as the downstream activation of various FSHD-associated pathways, which allowed us to correlate DUX4 expression signature dynamics with that of regulatory complexes, thereby facilitating the prioritization of epigenetic targets for DUX4 silencing. Single-cell transcriptomics combined with pseudotime modeling thus holds valuable information on FSHD disease etiology and progression that can potentially guide biomarker and target selection for therapy.

Michael Kyba - One of the best experts on this subject based on the ideXlab platform.

  • dna aptamers against the DUX4 protein reveal novel therapeutic implications for fshd
    The FASEB Journal, 2020
    Co-Authors: Christian Klingler, Michael Kyba, Ke Shi, Hideki Aihara, Jon Ashley, Adeline Stiefvater, Michael Sinnreich, Jochen Kinter
    Abstract:

    Aberrant expression of the transcription factor double homeobox protein 4 (DUX4) can lead to a number of diseases including facio-scapulo-humeral muscular dystrophy (FSHD), acute lymphoblastic leukemia, and sarcomas. Inhibition of DUX4 may represent a therapeutic strategy for these diseases. By applying Systematic Evolution of Ligands by EXponential Enrichment (SELEX), we identified aptamers against DUX4 with specific secondary structural elements conveying high affinity to DUX4 as assessed by fluorescence resonance energy transfer and fluorescence polarization techniques. Sequences analysis of these aptamers revealed the presence of two consensus DUX4 motifs in a reverse complementary fashion forming hairpins interspersed with bulge loops at distinct positions that enlarged the binding surface with the DUX4 protein, as determined by crystal structure analysis. We demonstrate that insertion of specific structural elements into transcription factor binding oligonucleotides can enhance specificity and affinity.

  • a novel p300 inhibitor reverses DUX4 mediated global histone h3 hyperacetylation target gene expression and cell death
    Science Advances, 2019
    Co-Authors: Darko Bosnakovski, Erik A. Toso, Meiricris T Silva, Sithara T Sunny, Elizabeth T Ener, Ce Yuan, Ziyou Cui, Michael A Walters, Ajit Jadhav, Michael Kyba
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) results from mutations causing overexpression of the transcription factor, DUX4, which interacts with the histone acetyltransferases, EP300 and CBP. We describe the activity of a new spirocyclic EP300/CBP inhibitor, iP300w, which inhibits the cytotoxicity of the DUX4 protein and reverses the overexpression of most DUX4 target genes, in engineered cell lines and FSHD myoblasts, as well as in an FSHD animal model. In evaluating the effect of iP300w on global histone H3 acetylation, we discovered that DUX4 overexpression leads to a dramatic global increase in the total amount of acetylated histone H3. This unexpected effect requires the C-terminus of DUX4, is conserved with mouse Dux, and may facilitate zygotic genome activation. This global increase in histone H3 acetylation is reversed by iP300w, highlighting the central role of EP300 and CBP in the transcriptional mechanism underlying DUX4 cytotoxicity and the translational potential of blocking this interaction.

  • crystal structure of the double homeodomain of DUX4 in complex with dna
    Cell Reports, 2018
    Co-Authors: John K. Lee, Michael Kyba, Darko Bosnakovski, Erik A. Toso, Tracy Dinh, Surajit Banerjee, Thomas E. Bohl, Ke Shi, Kayo Orellana, Hideki Aihara
    Abstract:

    Summary Double homeobox (DUX) transcription factors are unique to eutherian mammals. DUX4 regulates expression of repetitive elements during early embryogenesis, but misexpression of DUX4 causes facioscapulohumeral muscular dystrophy (FSHD) and translocations overexpressing the DUX4 double homeodomain cause B cell leukemia. Here, we report the crystal structure of the tandem homeodomains of DUX4 bound to DNA. The homeodomains bind DNA in a head-to-head fashion, with the linker making anchoring DNA minor-groove interactions and unique protein contacts. Remarkably, despite being tandem duplicates, the DUX4 homeodomains recognize different core sequences. This results from an arginine-to-glutamate mutation, unique to primates, causing alternative positioning of a key arginine side chain in the recognition helix. Mutational studies demonstrate that this primate-specific change is responsible for the divergence in sequence recognition that likely drove coevolution of embryonically regulated repeats in primates. Our work provides a framework for understanding the endogenous function of DUX4 and its role in FSHD and cancer.

  • low level DUX4 expression disrupts myogenesis through deregulation of myogenic gene expression
    Scientific Reports, 2018
    Co-Authors: Darko Bosnakovski, Erik A. Toso, Elizabeth T Ener, Micah D Gearhart, Si Ho Choi, Michael Kyba
    Abstract:

    Loss of silencing of the DUX4 gene on chromosome 4 causes facioscapulohumeral muscular dystrophy. While high level DUX4 expression induces apoptosis, the effects of low level DUX4 expression on human myogenic cells are not well understood. Low levels and sporadic expression of DUX4 have been reported in FSHD biopsy samples and myoblast cultures. Here, we show that a large set of human myogenic genes is rapidly deregulated by DUX4, including MYOD1 and MYF5, which are efficiently repressed even by low, non-toxic levels of DUX4. Human myoblasts modified to express low nontoxic levels of DUX4 were significantly impaired from differentiating into myotubes in vitro. Surprisingly, inhibition of differentiation does not require the transcriptional activation domain, thus is likely a feature of all mammalian DUX genes. DUX4 does not bind near the MYF5 gene, but has a prominent ChIP-seq peak within the MYF5 −118 kb enhancer. We find that when DUX4 binds at this site, it directs enhancer activity towards a nearby transcriptional start site for a noncoding nonfunctional RNA we name DIME (DUX4-induced MYF5 enhancer) transcript. These data highlight the anti-myogenic properties of DUX4 in human myogenic progenitor cells, and provide an example of enhancer disruption in the downregulation of MYF5.

  • Comment on structural basis of DUX4/IGH-driven transactivation
    2018
    Co-Authors: Hideki Aihara, John K. Lee, Darko Bosnakovski, Ke Shi, Michael Kyba
    Abstract:

    In a recent issue of Leukemia, Dong et al. reported the crystal structure of an isolated second homeodomain of human DUX4 (DUX4HD2) bound to a double-stranded DNA fragment (strand-1: 5’-TTCTAATCTAATCX-3’, X=A according to Materials and methods of the original paper[1], annealed with strand-2: 5’-AAGATTAGATTAGT-3’, whereas the sequence record of PDB entry 5z2t shows X=TT. We also note that 5’-TGATTAGATTAGAA-3’ in Materials and methods of the original paper is the reverse sequence of ‘strand-2’ above, which is from the PDB.) containing the sequence that authors refer to as the DUX4 responsive element (DRE), 5’-TAATCTAAT-3’ [1]. The reported structure consisted of two molecules of DUX4HD2 bound to DNA in a head-to-tail arrangement, with each DUX4HD2 recognizing a TAAT motif on the same strand. We were surprised by this configuration, because the DUX4 recognition sequence as defined by ChIP-seq[2, 3] and by mutagenesis[4] is actually 11-nucleotide (nt) long, not 9 nt, namely, 5’-TAATCTAATCA-3’. The terminal CA nucleotides are among the most highly conserved, but an explanation for their necessity is not provided by the structure presented. An additional curiosity was the fact that the apparently critical TAAT sequences are not present in the mouse Dux consensus sequence, which nevertheless has many residues in common with that of human DUX4, and also includes a strongly conserved CA at positions 10 and 11: 5’-TGATTCAATCA-3’ [5, 6]

Christopher R S Banerji - One of the best experts on this subject based on the ideXlab platform.

  • Lymphocytes contribute to DUX4 target genes in FSHD muscle biopsies
    2019
    Co-Authors: Christopher R S Banerji, Maryna Panamarova, Peter S Zammit
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) is an incurable myopathy linked to overexpression of DUX4. However, DUX4 is difficult to detect in FSHD myoblasts and target gene expression is not a consistent FSHD muscle biopsy biomarker, displaying efficacy only on pathologically inflamed samples. Immune gene misregulation occurs in FSHD muscle biopsies with DUX4 targets enriched for inflammatory processes. However, assessment of the FSHD immune cell transcriptome, and the evaluation of DUX4 and target gene expression has not yet been performed. We show that FSHD lymphoblastoid cell lines (LCLs) display robust DUX4 expression, and express early and late DUX4 targets. Moreover, genes elevated on FSHD LCLs are elevated in FSHD muscle biopsies, correlating with DUX4 target activation and histological inflammation. These genes are importantly unaltered in FSHD myoblasts/myotubes, implying a non-muscle source in biopsies. Our results indicate an immune cell source of DUX4 and target gene expression in FSHD muscle biopsies.

  • pax7 target gene repression is a superior fshd biomarker than DUX4 target gene activation associating with pathological severity and identifying fshd at the single cell level
    Human Molecular Genetics, 2019
    Co-Authors: Christopher R S Banerji, Peter S Zammit
    Abstract:

    : Facioscapulohumeral muscular dystrophy (FSHD) is a prevalent, incurable skeletal myopathy. The condition is linked to hypomethylation of the D4Z4 macrosatellite repeat at chromosome 4q35, leading to epigenetic derepression of the transcription factor DUX4; coupled with a permissive 4qA haplotype supplying a poly(A) signal. DUX4 may drive FSHD pathology via both induction of target genes and inhibition of the function of the myogenic master regulator PAX7. Biomarkers for FSHD have focused on DUX4 target gene expression. We have, however, reported that PAX7 target gene repression is a hallmark of FSHD skeletal muscle. Here we demonstrate that PAX7 target gene repression is an equivalent biomarker to DUX4 target gene expression when considering RNA-Sequencing data from magnetic resonance imaging-guided muscle biopsies. Moreover, PAX7 target gene repression correlates with active disease, independent to DUX4 target gene expression. PAX7 target genes are also repressed in RNA-Sequencing data from single cells, representing a significantly better biomarker of FSHD cells than DUX4 target gene expression. Importantly, PAX7 target gene repression is a significant biomarker in the majority of FSHD cells that are DUX4 target gene negative, and on which the DUX4 biomarker is indiscriminate. To facilitate the evaluation of validated biomarkers we provide a simple tool that outputs biomarker values from a normalized expression data matrix. In summary, PAX7 target gene repression in FSHD correlates with disease severity, independently of DUX4 target gene expression. At the single-cell level, PAX7 target gene repression can efficiently discriminate FSHD cells, even when no DUX4 target genes are detectable.

  • pax7 target genes are globally repressed in facioscapulohumeral muscular dystrophy skeletal muscle
    Nature Communications, 2017
    Co-Authors: Christopher R S Banerji, Simone Severini, Maryna Panamarova, Husam Hebaishi, Frederic Relaix, Robert B. White, Peter S Zammit
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) is a prevalent, incurable myopathy, linked to hypomethylation of D4Z4 repeats on chromosome 4q causing expression of the DUX4 transcription factor. However, DUX4 is difficult to detect in FSHD muscle biopsies and it is debatable how robust changes in DUX4 target gene expression are as an FSHD biomarker. PAX7 is a master regulator of myogenesis that rescues DUX4-mediated apoptosis. Here, we show that suppression of PAX7 target genes is a hallmark of FSHD, and that it is as major a signature of FSHD muscle as DUX4 target gene expression. This is shown using meta-analysis of over six FSHD muscle biopsy gene expression studies, and validated by RNA-sequencing on FSHD patient-derived myoblasts. DUX4 also inhibits PAX7 from activating its transcriptional target genes and vice versa. Furthermore, PAX7 target gene repression can explain oxidative stress sensitivity and epigenetic changes in FSHD. Thus, PAX7 target gene repression is a hallmark of FSHD that should be considered in the investigation of FSHD pathology and therapy. Facioscapulohumeral muscular dystrophy is a myopathy linked to ectopic expression of the DUX4 transcription factor. The authors show that the suppression of targets genes of the myogenesis regulator PAX7 is a signature of FSHD, and might explain oxidative stress sensitivity and epigenetic changes.

  • ret function in muscle stem cells points to tyrosine kinase inhibitor therapy for facioscapulohumeral muscular dystrophy
    eLife, 2016
    Co-Authors: Christopher R S Banerji, Louise A Moyle, Eric Blanc, Oihane Jaka, Johanna Prueller, Francesco Saverio Tedesco, Stephen D R Harridge, Rob Knight
    Abstract:

    Facioscapulohumeral muscular dystrophy (FSHD) involves sporadic expression of DUX4, which inhibits myogenesis and is pro-apoptotic. To identify target genes, we over-expressed DUX4 in myoblasts and found that the receptor tyrosine kinase Ret was significantly up-regulated, suggesting a role in FSHD. RET is dynamically expressed during myogenic progression in mouse and human myoblasts. Constitutive expression of either RET9 or RET51 increased myoblast proliferation, whereas siRNA-mediated knockdown of Ret induced myogenic differentiation. Suppressing RET activity using Sunitinib, a clinically-approved tyrosine kinase inhibitor, rescued differentiation in both DUX4-expressing murine myoblasts and in FSHD patient-derived myoblasts. Importantly, Sunitinib also increased engraftment and differentiation of FSHD myoblasts in regenerating mouse muscle. Thus, DUX4-mediated activation of Ret prevents myogenic differentiation and could contribute to FSHD pathology by preventing satellite cell-mediated repair. Rescue of DUX4-induced pathology by Sunitinib highlights the therapeutic potential of tyrosine kinase inhibitors for treatment of FSHD.

  • DUX4 induces a transcriptome more characteristic of a less differentiated cell state and inhibits myogenesis
    Journal of Cell Science, 2016
    Co-Authors: Paul Knopp, Christopher R S Banerji, Maryna Panamarova, Louise A Moyle, Yvonne D Krom, Bianca Den Hamer, Silvere M Van Der Maarel, Peter S Zammit
    Abstract:

    Skeletal muscle wasting in facioscapulohumeral muscular dystrophy (FSHD) results in substantial morbidity. On a disease-permissive chromosome 4qA haplotype, genomic and/or epigenetic changes at the D4Z4 macrosatellite repeat allows transcription of the DUX4 retrogene. Analysing transgenic mice carrying a human D4Z4 genomic locus from an FSHD-affected individual showed that DUX4 was transiently induced in myoblasts during skeletal muscle regeneration. Centromeric to the D4Z4 repeats is an inverted D4Z4 unit encoding DUX4c. Expression of DUX4, DUX4c and DUX4 constructs, including constitutively active, dominant-negative and truncated versions, revealed that DUX4 activates target genes to inhibit proliferation and differentiation of satellite cells, but that it also downregulates target genes to suppress myogenic differentiation. These transcriptional changes elicited by DUX4 in mouse have significant overlap with genes regulated by DUX4 in man. Comparison of DUX4 and DUX4c transcriptional perturbations revealed that DUX4 regulates genes involved in cell proliferation, whereas DUX4c regulates genes engaged in angiogenesis and muscle development, with both DUX4 and DUX4c modifing genes involved in urogenital development. Transcriptomic analysis showed that DUX4 operates through both target gene activation and repression to orchestrate a transcriptome characteristic of a less-differentiated cell state.

Sean C Shadle - One of the best experts on this subject based on the ideXlab platform.

  • p53 convergently activates dux DUX4 in embryonic stem cells and in facioscapulohumeral muscular dystrophy cell models
    Nature Genetics, 2021
    Co-Authors: Edward J Grow, Peter G Hendrickson, Bradley D Weaver, Sean C Shadle, Christina M Smith, Jingtao Guo, Paula Stein, Nicholas E Johnson, Russell J Butterfield, Roberta Menafra
    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.

  • DUX4 induced bidirectional hsatii satellite repeat transcripts form intranuclear double stranded rna foci in human cell models of fshd
    Human Molecular Genetics, 2019
    Co-Authors: Chao-jen Wong, Sean R. Bennett, Silvere M Van Der Maarel, Amy E Campbell, Sean C Shadle, Nancy A Karreman, Brenda L Bass, Stephen J. Tapscott
    Abstract:

    : The DUX4 transcription factor is normally expressed in the cleavage stage embryo and regulates genes involved in embryonic genome activation. Mis-expression 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.

  • DUX4 suppresses mhc class i to promote cancer immune evasion and resistance to checkpoint blockade
    Developmental Cell, 2019
    Co-Authors: Guoliang Chew, Stephen J. Tapscott, Amy E Campbell, Sean C Shadle, Emma J De Neef, Nicholas A Sutliff, Robert K Bradley
    Abstract:

    Summary 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.

  • nurd and caf 1 mediated silencing of the d4z4 array is modulated by DUX4 induced mbd3l proteins
    eLife, 2018
    Co-Authors: Amy E Campbell, Rebecca Resnick, Silvère M. Van Der Maarel, Rabi Tawil, Jongwon Lim, Sean C Shadle, Sujatha Jagannathan, Stephen J. Tapscott
    Abstract:

    The DNA sequences of humans and other mammals contain many repetitive regions. This repetition makes these regions difficult to study with conventional approaches, and so the exact role of repetitive DNA is not fully understood. A particular sequence of repetitive DNA that plays an important role in human health contains a gene called DUX4 in each repeat. DUX4 is normally active in stem cells and in early-stage embryos. This gene is then switched off or ‘silenced’ during later stages of development and in most cells of the body. However, in some individuals the DUX4 gene inappropriately activates in muscle cells. This causes a disease known as facioscapulohumeral muscular dystrophy (FSHD), in which muscle weakness begins in the face and upper body and eventually spreads to other muscles. Currently, there is no cure for FSHD. Proteins that bind to DNA can control the activity of nearby genes. Little is known about which proteins silence DUX4 at the appropriate time and in the right cells, so Campbell et al. set out to identify the proteins that attach to the repetitive DNA sequences containing DUX4. Further investigation showed that several of these proteins play an important role in keeping DUX4 turned off, including two protein complexes called NuRD and CAF-1. These complexes are necessary to silence DUX4 in human muscle cells and stem cells. Campbell et al. also identified a protein that can increase the activity of the DUX4 gene in FSHD muscle cells by overcoming the silencing activity of the NuRD complex. Overall, the results presented by Campbell et al. provide the groundwork for developing new treatments for FSHD. The next step will be to discover ways of enhancing the ability of NuRD and CAF-1 to silence the DUX4 gene.

  • bet bromodomain inhibitors and agonists of the beta 2 adrenergic receptor identified in screens for compounds that inhibit DUX4 expression in fshd muscle cells
    Skeletal Muscle, 2017
    Co-Authors: Amy E Campbell, Lauren Snider, Jonathan Oliva, Matthew P Yates, Jun Wen Zhong, Sean C Shadle, Nikita Singh, Shannon Tai, Yosuke Hiramuki
    Abstract:

    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. 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. 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. 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.