Bone morphogenetic protein 4 reduces global H3K4me3 to inhibit proliferation and promote differentiation of human neural stem cells

Posttranslational modifications (PTMs) on histone tails spatiotemporally dictate mammalian neural stem cell (NSC) fate. Bone morphogenetic protein 4 (BMP4), a member of the transforming growth factor β (TGF-β) superfamily, suppresses NSC proliferation and fosters differentiation into astroglial cells. Whether PTMs mediate these effects of BMP4 is unknown. Here we demonstrate that BMP4 signaling causes a net reduction in cellular histone H3 lysine 4 trimethylation (H3K4me3), an active histone mark at promoters of genes associated with human NSC proliferation. We also show that H3K4me3 reduction by BMP4 is mediated by decreased expression of SETD1A and WDR82, two methyltransferase components of SETD1A-COMPASS. Down-regulation of these components decreases expression of key genes expressed in hNSCs, while ectopic expression via transfection dedifferentiates human astrocytes (HAs). These observations suggest that BMP4 influences NSC fate by regulating PTMs and altering chromatin structure. SIGNIFICANCE STATEMENT BMP4 is critical in determining hNSC fate. Whether histone posttranslational modifications (PTM) mediate the effects of BMP4 is unknown. Here we report that H3K4me3, brought about by its methyltransferases SETD1A and WDR82, at promoters of stem cell genes OCT4 and NESTIN is involved in human neural stem cell (hNSC) maintenance. BMP4 promotes hNSC astroglial differentiation in part through reduction of SETD1A and WDR82 and thus decreased frequency of H3K4me3 at the promoters of these genes. These results provide evidence that BMP4 promotes hNSC differentiation through a potential epigenetic mechanism and extend our understanding of the role of histone PTM in central nervous system development.

[1]  S. Armstrong,et al.  A Non-catalytic Function of SETD1A Regulates Cyclin K and the DNA Damage Response , 2018, Cell.

[2]  C. James,et al.  Therapeutic Potential for Bone Morphogenetic Protein 4 in Human Malignant Glioma , 2017, Neoplasia.

[3]  Yong Zhang,et al.  H3K4 Methyltransferase Set1a Is A Key Oct4 Coactivator Essential for Generation of Oct4 Positive Inner Cell Mass , 2016, Stem cells.

[4]  S. Maheswaran,et al.  SETD1A induced miRNA network suppresses the p53 gene expression module , 2016, Cell cycle.

[5]  Aaron C. Daugherty,et al.  H3K4me3 Breadth Is Linked to Cell Identity and Transcriptional Consistency , 2014, Cell.

[6]  Ben S. Wittner,et al.  Supplementary Information Supplementary Figures , 2022 .

[7]  C. James,et al.  Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma , 2014, Nature Medicine.

[8]  Aaron C. Daugherty,et al.  H3K4me3 Breadth Is Linked to Cell Identity and Transcriptional Consistency , 2014, Cell.

[9]  Chin-Hsing Annie Lin,et al.  Epigenetic Regulation by Chromatin Activation Mark H3K4me3 in Primate Progenitor Cells within Adult Neurogenic Niche , 2014, Scientific Reports.

[10]  A. F. Stewart,et al.  The H3K4 methyltransferase Setd1a is first required at the epiblast stage, whereas Setd1b becomes essential after gastrulation , 2014, Development.

[11]  Min Gyu Lee,et al.  Histone H3 lysine 4 methyltransferases and demethylases in self-renewal and differentiation of stem cells , 2013, Cell & Bioscience.

[12]  Sabine Mueller,et al.  The histone H3.3K27M mutation in pediatric glioma reprograms H3K27 methylation and gene expression. , 2013, Genes & development.

[13]  R. Chen,et al.  The Trithorax Group Protein Ash2l Is Essential for Pluripotency and Maintaining Open Chromatin in Embryonic Stem Cells* , 2012, The Journal of Biological Chemistry.

[14]  V. Coskun,et al.  Epigenetic regulation of stem cells differentiating along the neural lineage , 2012, Current Opinion in Neurobiology.

[15]  S. Hung,et al.  Oct4 and Nanog directly regulate Dnmt1 to maintain self-renewal and undifferentiated state in mesenchymal stem cells. , 2012, Molecular cell.

[16]  J. Kessler,et al.  The dynamic role of bone morphogenetic proteins in neural stem cell fate and maturation , 2012, Developmental neurobiology.

[17]  A. Shilatifard The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. , 2012, Annual review of biochemistry.

[18]  A. Basu,et al.  Epigenetic regulation of self‐renewal and fate determination in neural stem cells , 2012, Journal of neuroscience research.

[19]  N. Tommerup,et al.  Jarid1b targets genes regulating development and is involved in neural differentiation , 2011, The EMBO journal.

[20]  Suk-Ho Lee,et al.  Prolonged Membrane Depolarization Enhances Midbrain Dopamine Neuron Differentiation via Epigenetic Histone Modifications , 2011, Stem cells.

[21]  G. Ming,et al.  Adult Neurogenesis in the Mammalian Brain: Significant Answers and Significant Questions , 2011, Neuron.

[22]  Jonathan M. Monk,et al.  Wdr5 Mediates Self-Renewal and Reprogramming via the Embryonic Stem Cell Core Transcriptional Network , 2011, Cell.

[23]  Qi Zhou,et al.  WDR82, a Key Epigenetics-Related Factor, Plays a Crucial Role in Normal Early Embryonic Development in Mice1 , 2011, Biology of reproduction.

[24]  Y. Pu,et al.  Bone morphogenetic protein 4 inhibits cell proliferation and induces apoptosis in glioma stem cells. , 2011, Cancer biotherapy & radiopharmaceuticals.

[25]  B. Bernstein,et al.  Role for Dpy-30 in ES Cell-Fate Specification by Regulation of H3K4 Methylation within Bivalent Domains , 2011, Cell.

[26]  K. Nakashima,et al.  Chromatin remodeling in neural stem cell differentiation , 2010, Current Opinion in Neurobiology.

[27]  K. Nakashima,et al.  Epigenetic regulation in neural stem cell differentiation , 2010, Development, growth & differentiation.

[28]  M. Doughty,et al.  Role of H3K4 demethylases in complex neurodevelopmental diseases. , 2010, Epigenomics.

[29]  J. García-Verdugo,et al.  Chromatin remodelling factor Mll1 is essential for neurogenesis from postnatal neural stem cells , 2009, Nature.

[30]  O. Hermanson,et al.  CXXC5 Is a Novel BMP4-regulated Modulator of Wnt Signaling in Neural Stem Cells* , 2009, Journal of Biological Chemistry.

[31]  Peng Fei Wang,et al.  Molecular Regulation of H3K4 Trimethylation by Wdr82, a Component of Human Set1/COMPASS , 2008, Molecular and Cellular Biology.

[32]  F. Gage,et al.  Mechanisms and Functional Implications of Adult Neurogenesis , 2008, Cell.

[33]  D. Skalnik,et al.  Wdr82 Is a C-Terminal Domain-Binding Protein That Recruits the Setd1A Histone H3-Lys4 Methyltransferase Complex to Transcription Start Sites of Transcribed Human Genes , 2007, Molecular and Cellular Biology.

[34]  A. Shilatifard,et al.  Covalent modifications of histones during development and disease pathogenesis , 2007, Nature Structural &Molecular Biology.

[35]  John T. Dimos,et al.  shRNA knockdown of Bmi-1 reveals a critical role for p21-Rb pathway in NSC self-renewal during development. , 2007, Cell stem cell.

[36]  D. Skalnik,et al.  Identification and Characterization of the Human Set1B Histone H3-Lys4 Methyltransferase Complex* , 2007, Journal of Biological Chemistry.

[37]  D. Panchision,et al.  Concise Review: Bone Morphogenetic Protein Pleiotropism in Neural Stem Cells and Their Derivatives—Alternative Pathways, Convergent Signals , 2007, Stem cells.

[38]  G. Broggi,et al.  Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells , 2006, Nature.

[39]  A. F. Stewart,et al.  Multiple epigenetic maintenance factors implicated by the loss of Mll2 in mouse development , 2006, Development.

[40]  Eric S. Lander,et al.  Genomic Maps and Comparative Analysis of Histone Modifications in Human and Mouse , 2005, Cell.

[41]  J. Kessler,et al.  Interactions between ID and OLIG proteins mediate the inhibitory effects of BMP4 on oligodendroglial differentiation , 2004, Development.

[42]  Jing Wang,et al.  BMP4 supports self-renewal of embryonic stem cells by inhibiting mitogen-activated protein kinase pathways. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[43]  S. Khorasanizadeh The Nucleosome From Genomic Organization to Genomic Regulation , 2004, Cell.

[44]  J. Kessler,et al.  Msx2 and p21CIP1/WAF1 mediate the proapoptotic effects of bone morphogenetic protein‐4 on ventricular zone progenitor cells , 2002, Journal of neuroscience research.

[45]  R. McKay,et al.  The control of neural stem cells by morphogenic signals. , 2002, Current opinion in genetics & development.

[46]  J. Loturco,et al.  Neuronal Differentiation of Precursors in the Neocortical Ventricular Zone Is Triggered by BMP , 1998, The Journal of Neuroscience.

[47]  M. Rao,et al.  Spinal Cord Neuronal Precursors Generate Multiple Neuronal Phenotypes in Culture , 1998, The Journal of Neuroscience.

[48]  B. Bernstein,et al.  Charting histone modifications and the functional organization of mammalian genomes , 2011, Nature Reviews Genetics.

[49]  K. Xia,et al.  Genome-wide mapping of SMAD target genes reveals the role of BMP signaling in embryonic stem cell fate determination. , 2010, Genome research.

[50]  Tony Kouzarides,et al.  Histone H3 lysine 4 methylation patterns in higher eukaryotic genes , 2004, Nature Cell Biology.