The myopathic transcription factor DUX4 induces the production of truncated RNA-binding proteins in human muscle cells

DUX4 is an embryonic transcription factor whose misexpression in skeletal muscle causes facioscapulohumeral muscular dystrophy (FSHD). DUX4 induces the transcription of thousands of RNAs and dysregulates multiple pathways that could contribute to FSHD pathophysiology. However, lack of temporal data and the knowledge of which RNAs are actively translated following DUX4 expression has hindered our understanding of the cascade of events that lead to muscle cell death. Here, we interrogate the DUX4 transcriptome and translatome over time and find dysregulation of most key pathways as early as 4 hours after DUX4 induction, demonstrating the potent effect of DUX4 in disrupting muscle biology. We also observe extensive transcript downregulation as well as induction, and a high concordance between mRNA abundance and translation status. Significantly, DUX4 triggers widespread production of truncated protein products derived from aberrant RNAs that are degraded in normal muscle cells. One such protein, truncated serine/arginine-rich splicing factor 3 (SRSF3-TR), is present in FSHD muscle cells and disrupts splicing autoregulation when ectopically expressed in myoblasts. Taken together, the temporal dynamics of DUX4 induction show how the pathologic presence of an embryonic transcription factor in muscle cells alters gene expression to ultimately perturb RNA homeostasis.

[1]  N. Geijsen,et al.  DUX4 induces a homogeneous sequence of molecular changes, culminating in the activation of a stem-cell-like transcriptional network and induction of apoptosis in somatic cells , 2021, bioRxiv.

[2]  J. George,et al.  Poison Exon Splicing Regulates a Coordinated Network of SR Protein Expression during Differentiation and Tumorigenesis. , 2020, Molecular cell.

[3]  Peifeng Li,et al.  Emerging Roles of SRSF3 as a Therapeutic Target for Cancer , 2020, Frontiers in Oncology.

[4]  Arun Kumar,et al.  SRSF3: Newly discovered functions and roles in human health and diseases. , 2020, European journal of cell biology.

[5]  Neville E. Sanjana,et al.  Applying genome-wide CRISPR-Cas9 screens for therapeutic discovery in facioscapulohumeral muscular dystrophy , 2020, Science Translational Medicine.

[6]  Marcel H. Schulz,et al.  SRSF7 maintains its homeostasis through the expression of Split-ORFs and nuclear body assembly , 2020, Nature Structural & Molecular Biology.

[7]  K. Lim,et al.  DUX4 Signalling in the Pathogenesis of Facioscapulohumeral Muscular Dystrophy , 2020, International journal of molecular sciences.

[8]  J. Leszyk,et al.  Identification of the hyaluronic acid pathway as a therapeutic target for facioscapulohumeral muscular dystrophy , 2019, Science Advances.

[9]  S. Henikoff,et al.  DUX4-Induced Histone Variants H3.X and H3.Y Mark DUX4 Target Genes for Expression , 2019, Cell reports.

[10]  John F Ouyang,et al.  deltaTE: Detection of Translationally Regulated Genes by Integrative Analysis of Ribo‐seq and RNA‐seq Data , 2019, Current protocols in molecular biology.

[11]  M. Kyba,et al.  A novel P300 inhibitor reverses DUX4-mediated global histone H3 hyperacetylation, target gene expression, and cell death , 2019, Science Advances.

[12]  S. Tapscott,et al.  DUX4 Suppresses MHC Class I to Promote Cancer Immune Evasion and Resistance to Checkpoint Blockade. , 2019, Developmental cell.

[13]  S. Tapscott,et al.  DUX4-induced bidirectional HSATII satellite repeat transcripts form intranuclear double stranded RNA foci in human cell models of FSHD , 2019, bioRxiv.

[14]  Michaela Müller-McNicoll,et al.  Auto-regulatory feedback by RNA-binding proteins , 2019, Journal of molecular cell biology.

[15]  U. Ohler,et al.  Quantification of translation uncovers the functions of the alternative transcriptome , 2019, bioRxiv.

[16]  U. Ohler,et al.  Ribo-seQC: comprehensive analysis of cytoplasmic and organellar ribosome profiling data , 2019, bioRxiv.

[17]  M. Landthaler,et al.  DDX3 depletion represses translation of mRNAs with complex 5′ UTRs , 2019, bioRxiv.

[18]  L. Chee,et al.  Pan-Cancer Analyses Reveal Genomic Features of FOXM1 Overexpression in Cancer , 2019, Cancers.

[19]  Philip R. Gafken,et al.  Quantitative proteomics reveals key roles for post-transcriptional gene regulation in the molecular pathology of facioscapulohumeral muscular dystrophy , 2019, eLife.

[20]  M. Kyba,et al.  Low level DUX4 expression disrupts myogenesis through deregulation of myogenic gene expression , 2018, Scientific Reports.

[21]  R. Tawil,et al.  Facioscapulohumeral Muscular Dystrophy: Update on Pathogenesis and Future Treatments , 2018, Neurotherapeutics.

[22]  S. Tapscott,et al.  Facioscapulohumeral dystrophy: activating an early embryonic transcriptional program in human skeletal muscle. , 2018, Human molecular genetics.

[23]  Joseph G Ibrahim,et al.  Heavy-tailed prior distributions for sequence count data: removing the noise and preserving large differences , 2018, bioRxiv.

[24]  Simone Severini,et al.  PAX7 target genes are globally repressed in facioscapulohumeral muscular dystrophy skeletal muscle , 2017, Nature Communications.

[25]  M. Kyba,et al.  The DUX4 homeodomains mediate inhibition of myogenesis and are functionally exchangeable with the Pax7 homeodomain , 2017, Journal of Cell Science.

[26]  Michael Kyba,et al.  Muscle pathology from stochastic low level DUX4 expression in an FSHD mouse model , 2017, Nature Communications.

[27]  Stephen J. Tapscott,et al.  Conservation and innovation in the DUX4-family gene network , 2017, Nature Genetics.

[28]  S. Tapscott,et al.  DUX4-induced dsRNA and MYC mRNA stabilization activate apoptotic pathways in human cell models of facioscapulohumeral dystrophy , 2017, PLoS genetics.

[29]  Yi-Wen Chen,et al.  DUX 4 differentially regulates transcriptomes of human rhabdomyosarcoma and mouse C 2 C 12 cells , 2017 .

[30]  F. Boyce,et al.  Nuclear bodies reorganize during myogenesis in vitro and are differentially disrupted by expression of FSHD-associated DUX4 , 2016, Skeletal Muscle.

[31]  S. Tapscott,et al.  Model systems of DUX4 expression recapitulate the transcriptional profile of FSHD cells. , 2016, Human molecular genetics.

[32]  L. Maquat,et al.  Nonsense-mediated mRNA decay in humans at a glance , 2016, Journal of Cell Science.

[33]  Daniel G. Miller,et al.  Endogenous DUX4 expression in FSHD myotubes is sufficient to cause cell death and disrupts RNA splicing and cell migration pathways. , 2015, Human molecular genetics.

[34]  Raphael Gottardo,et al.  Orchestrating high-throughput genomic analysis with Bioconductor , 2015, Nature Methods.

[35]  F. Boyce,et al.  Expression of FSHD-related DUX4-FL alters proteostasis and induces TDP-43 aggregation , 2015, Annals of clinical and translational neurology.

[36]  S. Tapscott,et al.  A feedback loop between nonsense-mediated decay and the retrogene DUX4 in facioscapulohumeral muscular dystrophy , 2015, eLife.

[37]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[38]  Zizhen Yao,et al.  DUX4-induced gene expression is the major molecular signature in FSHD skeletal muscle. , 2014, Human molecular genetics.

[39]  M. Kyba,et al.  Dominant lethal pathologies in male mice engineered to contain an X-linked DUX4 transgene. , 2014, Cell reports.

[40]  S. Tapscott,et al.  Facioscapulohumeral dystrophy: the path to consensus on pathophysiology , 2014, Skeletal Muscle.

[41]  Nicholas T. Ingolia Ribosome profiling: new views of translation, from single codons to genome scale , 2014, Nature Reviews Genetics.

[42]  Wei Shi,et al.  featureCounts: an efficient general purpose program for assigning sequence reads to genomic features , 2013, Bioinform..

[43]  Yi-Wen Chen,et al.  DUX4 Differentially Regulates Transcriptomes of Human Rhabdomyosarcoma and Mouse C2C12 Cells , 2013, PloS one.

[44]  Eytan Domany,et al.  An accessible database for mouse and human whole transcriptome qPCR primers , 2013, Bioinform..

[45]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[46]  Kevin W Eliceiri,et al.  NIH Image to ImageJ: 25 years of image analysis , 2012, Nature Methods.

[47]  W. Huber,et al.  Detecting differential usage of exons from RNA-seq data , 2012, Genome research.

[48]  Steven L Salzberg,et al.  Fast gapped-read alignment with Bowtie 2 , 2012, Nature Methods.

[49]  Abraham P. Fong,et al.  DUX4 activates germline genes, retroelements, and immune mediators: implications for facioscapulohumeral dystrophy. , 2012, Developmental cell.

[50]  S. Harper,et al.  DUX4, a candidate gene for facioscapulohumeral muscular dystrophy, causes p53‐dependent myopathy in vivo , 2011, Annals of neurology.

[51]  Daniel G. Miller,et al.  A Unifying Genetic Model for Facioscapulohumeral Muscular Dystrophy , 2010, Science.

[52]  Hadley Wickham,et al.  ggplot2 - Elegant Graphics for Data Analysis (2nd Edition) , 2017 .

[53]  Michael Kyba,et al.  Biphasic Myopathic Phenotype of Mouse DUX, an ORF within Conserved FSHD-Related Repeats , 2009, PloS one.

[54]  M. Kyba,et al.  An isogenetic myoblast expression screen identifies DUX4‐mediated FSHD‐associated molecular pathologies , 2008, The EMBO journal.

[55]  M. Holcik,et al.  IRES-Mediated Translation of Utrophin A Is Enhanced by Glucocorticoid Treatment in Skeletal Muscle Cells , 2008, PloS one.

[56]  Chao Zhang,et al.  IRE1 Signaling Affects Cell Fate During the Unfolded Protein Response , 2007, Science.

[57]  A. Rosa,et al.  The DUX4 gene at the FSHD1A locus encodes a pro-apoptotic protein , 2007, Neuromuscular Disorders.

[58]  C. Proud eIF2 and the control of cell physiology. , 2005, Seminars in cell & developmental biology.

[59]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[60]  W. Merrick,et al.  Analysis of Eukaryotic Translation in Purified and Semipurified Systems , 2000, Current protocols in cell biology.

[61]  H. Jumaa,et al.  The splicing factor SRp20 modifies splicing of its own mRNA and ASF/SF2 antagonizes this regulation , 1997, The EMBO journal.