Hdac4 Interactions in Huntington's Disease Viewed Through the Prism of Multiomics*

The histone deacetylase Hdac4 is known to contribute to the progression of Huntington's Disease (HD), but the underlying mechanisms remain unknown. Here, we defined the endogenous interactome of Hdac4 in the brain of HD mouse models, characterizing their polyQ- and age-dependence in affected tissues. Further integration with proteome and transcriptome data sets reveals the disease-induced enhancement of Hdac4 interactions with vesicular sorting proteins, including the WASH complex. This may contribute to the known decreased synaptic functions in Huntington's Disease. Graphical Abstract Highlights Endogenous interactomes of Hdac4 and Hdac5 in mouse whole brain and striata. Hdac4 exhibits polyQ- and age-dependent interactions in Huntington's Disease models. Hdac4 associates with vesicular trafficking proteins, including the WASH complex. Multiomics analysis supports functional Hdac4 interactions in Huntington's Disease. Huntington's disease (HD) is a monogenic disorder, driven by the expansion of a trinucleotide (CAG) repeat within the huntingtin (Htt) gene and culminating in neuronal degeneration in the brain, predominantly in the striatum and cortex. Histone deacetylase 4 (Hdac4) was previously found to contribute to the disease progression, providing a potential therapeutic target. Hdac4 knockdown reduced accumulation of misfolded Htt protein and improved HD phenotypes. However, the underlying mechanism remains unclear, given its independence on deacetylase activity and the predominant cytoplasmic Hdac4 localization in the brain. Here, we undertook a multiomics approach to uncover the function of Hdac4 in the context of HD pathogenesis. We characterized the interactome of endogenous Hdac4 in brains of HD mouse models. Alterations in interactions were investigated in response to Htt polyQ length, comparing mice with normal (Q20) and disease (Q140) Htt, at both pre- and post-symptomatic ages (2 and 10 months, respectively). Parallel analyses for Hdac5, a related class IIa Hdac, highlighted the unique interaction network established by Hdac4. To validate and distinguish interactions specifically enhanced in an HD-vulnerable brain region, we next characterized endogenous Hdac4 interactions in dissected striata from this HD mouse series. Hdac4 associations were polyQ-dependent in the striatum, but not in the whole brain, particularly in symptomatic mice. Hdac5 interactions did not exhibit polyQ dependence. To identify which Hdac4 interactions and functions could participate in HD pathogenesis, we integrated our interactome with proteome and transcriptome data sets generated from the striata. We discovered an overlap in enriched functional classes with the Hdac4 interactome, particularly in vesicular trafficking and synaptic functions, and we further validated the Hdac4 interaction with the Wiskott-Aldrich Syndrome Protein and SCAR Homolog (WASH) complex. This study expands the knowledge of Hdac4 regulation and functions in HD, adding to the understanding of the molecular underpinning of HD phenotypes.

[1]  V. Mattis,et al.  Human Huntington's Disease iPSC-Derived Cortical Neurons Display Altered Transcriptomics, Morphology, and Maturation. , 2018, Cell reports.

[2]  Julia M. Schulze,et al.  Reading chromatin , 2010, Epigenetics.

[3]  D. Billadeau,et al.  A FAM21-containing WASH complex regulates retromer-dependent sorting. , 2009, Developmental cell.

[4]  J. Workman,et al.  ATAC is a double histone acetyltransferase complex that stimulates nucleosome sliding , 2008, Nature Structural &Molecular Biology.

[5]  Joseph A. Loo,et al.  Enhanced FASP (eFASP) to Increase Proteome Coverage and Sample Recovery for Quantitative Proteomic Experiments , 2014, Journal of proteome research.

[6]  Michael Q. Zhang,et al.  Purification and characterization of human RNPS1: a general activator of pre‐mRNA splicing , 1999, The EMBO journal.

[7]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[8]  Z. Weng,et al.  RNA Sequence Analysis of Human Huntington Disease Brain Reveals an Extensive Increase in Inflammatory and Developmental Gene Expression , 2015, PloS one.

[9]  G P Bates,et al.  Self-assembly of polyglutamine-containing huntingtin fragments into amyloid-like fibrils: implications for Huntington's disease pathology. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[10]  Eric H Kim,et al.  The Neuropathology of Huntington's Disease. , 2015, Current topics in behavioral neurosciences.

[11]  K. Shirahige,et al.  Decreased cohesin in the brain leads to defective synapse development and anxiety-related behavior , 2017, The Journal of experimental medicine.

[12]  M. MacDonald,et al.  HD CAG-correlated gene expression changes support a simple dominant gain of function. , 2011, Human molecular genetics.

[13]  S. Folstein,et al.  Corticotropin-releasing hormone (CRH) is decreased in the basal ganglia in Huntington's disease , 1987, Brain Research.

[14]  Dimitri Krainc,et al.  Transcriptional Repression of PGC-1α by Mutant Huntingtin Leads to Mitochondrial Dysfunction and Neurodegeneration , 2006, Cell.

[15]  Lei Zeng,et al.  Structure and ligand of a histone acetyltransferase bromodomain , 1999, Nature.

[16]  Amber L. Couzens,et al.  The CRAPome: a Contaminant Repository for Affinity Purification Mass Spectrometry Data , 2013, Nature Methods.

[17]  D. Housman,et al.  The Huntington's disease protein interacts with p53 and CREB-binding protein and represses transcription. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[18]  G. Bates,et al.  Histone deacetylase inhibitors as therapeutics for polyglutamine disorders , 2006, Nature Reviews Neuroscience.

[19]  A. F. Neuwald,et al.  HEAT repeats associated with condensins, cohesins, and other complexes involved in chromosome-related functions. , 2000, Genome research.

[20]  P. Marks,et al.  SAHA Decreases HDAC 2 and 4 Levels In Vivo and Improves Molecular Phenotypes in the R6/2 Mouse Model of Huntington's Disease , 2011, PloS one.

[21]  P. McColgan,et al.  Huntington's disease: a clinical review , 2018, European journal of neurology.

[22]  E. Olson,et al.  The many roles of histone deacetylases in development and physiology: implications for disease and therapy , 2009, Nature Reviews Genetics.

[23]  C. Ouimet,et al.  Regional and subcellular distribution of HDAC4 in mouse brain , 2010, The Journal of comparative neurology.

[24]  M. MacDonald,et al.  Trinucleotide repeat length and progression of illness in Huntington's disease. , 1994, Journal of medical genetics.

[25]  Haibei Hu,et al.  Mutant huntingtin affects the rate of transcription of striatum‐specific isoforms of phosphodiesterase 10A , 2004, The European journal of neuroscience.

[26]  Jeffrey R. Whiteaker,et al.  Proteogenomic characterization of human colon and rectal cancer , 2014, Nature.

[27]  Haibei Hu,et al.  Structure, expression and regulation of the cannabinoid receptor gene (CB1) in Huntington's disease transgenic mice. , 2004, European journal of biochemistry.

[28]  T. Dobránsky,et al.  N-type Ca2+ channels are affected by full-length mutant huntingtin expression in a mouse model of Huntington's disease , 2017, Neurobiology of Aging.

[29]  C. Blackstone,et al.  FAM21 directs SNX27–retromer cargoes to the plasma membrane by preventing transport to the Golgi apparatus , 2016, Nature Communications.

[30]  M. Seaman,et al.  Analysis of the Retromer complex-WASH complex interaction illuminates new avenues to explore in Parkinson disease , 2014, Communicative & integrative biology.

[31]  Song Tan,et al.  Structural and Functional Conservation of the NuA4 Histone Acetyltransferase Complex from Yeast to Humans , 2004, Molecular and Cellular Biology.

[32]  Amanda J. Guise,et al.  The functional interactome landscape of the human histone deacetylase family , 2013, Molecular systems biology.

[33]  Amanda J. Guise,et al.  Nuclear Import of Histone Deacetylase 5 by Requisite Nuclear Localization Signal Phosphorylation* , 2010, Molecular & Cellular Proteomics.

[34]  Z. Otwinowski,et al.  WASH and WAVE actin regulators of the Wiskott–Aldrich syndrome protein (WASP) family are controlled by analogous structurally related complexes , 2010, Proceedings of the National Academy of Sciences.

[35]  Hyungwon Choi,et al.  SAINT: Probabilistic Scoring of Affinity Purification - Mass Spectrometry Data , 2010, Nature Methods.

[36]  L. Smeeth,et al.  Prevalence of adult Huntington's disease in the UK based on diagnoses recorded in general practice records , 2013, Journal of Neurology, Neurosurgery & Psychiatry.

[37]  M. Mann,et al.  Universal sample preparation method for proteome analysis , 2009, Nature Methods.

[38]  Yujin E. Kim,et al.  Soluble Oligomers of PolyQ-Expanded Huntingtin Target a Multiplicity of Key Cellular Factors. , 2016, Molecular cell.

[39]  Erich E. Wanker,et al.  HDAC4 Reduction: A Novel Therapeutic Strategy to Target Cytoplasmic Huntingtin and Ameliorate Neurodegeneration , 2013, PLoS biology.

[40]  Matthias Mann,et al.  Cell type– and brain region–resolved mouse brain proteome , 2015, Nature Neuroscience.

[41]  R. Wetzel,et al.  Serine phosphorylation suppresses huntingtin amyloid accumulation by altering protein aggregation properties. , 2012, Journal of molecular biology.

[42]  Damian Szklarczyk,et al.  The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible , 2016, Nucleic Acids Res..

[43]  C. Ross,et al.  Huntington's disease: from molecular pathogenesis to clinical treatment , 2011, The Lancet Neurology.

[44]  S. Luo,et al.  Targeting Gpr52 lowers mutant HTT levels and rescues Huntington’s disease-associated phenotypes , 2018, Brain : a journal of neurology.

[45]  L. Raymond Striatal synaptic dysfunction and altered calcium regulation in Huntington disease. , 2017, Biochemical and biophysical research communications.

[46]  Giovanni Coppola,et al.  Integrated genomics and proteomics to define huntingtin CAG length-dependent networks in HD Mice , 2016, Nature Neuroscience.

[47]  S. D’Mello,et al.  Reduced Expression of Foxp1 as a Contributing Factor in Huntington's Disease , 2017, The Journal of Neuroscience.

[48]  L. M. Valor,et al.  Early alteration of epigenetic-related transcription in Huntington’s disease mouse models , 2018, Scientific Reports.

[49]  Ileana M. Cristea,et al.  Proteomics-based methods for discovery, quantification, and validation of protein-protein interactions. , 2013, Analytical chemistry.

[50]  J. Botas,et al.  A striatal-enriched intronic GPCR modulates huntingtin levels and toxicity , 2015, eLife.

[51]  V. Haucke,et al.  Phosphatidylinositol 3‐phosphates—at the interface between cell signalling and membrane traffic , 2016, The EMBO journal.

[52]  M. Zatz,et al.  Mutations in the KIAA0196 gene at the SPG8 locus cause hereditary spastic paraplegia. , 2007, American journal of human genetics.

[53]  Manish S. Shah,et al.  A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes , 1993, Cell.

[54]  Stefka Tyanova,et al.  Perseus: A Bioinformatics Platform for Integrative Analysis of Proteomics Data in Cancer Research. , 2018, Methods in molecular biology.

[55]  Yi Xing,et al.  Transcriptome sequencing reveals aberrant alternative splicing in Huntington's disease. , 2016, Human molecular genetics.

[56]  K. Nasmyth,et al.  Chromosomal Cohesin Forms a Ring , 2003, Cell.

[57]  S. W. Davies,et al.  Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain. , 1997, Science.

[58]  R. Kiyama,et al.  Kank proteins: a new family of ankyrin-repeat domain-containing proteins. , 2008, Biochimica et biophysica acta.

[59]  R. Taliyan,et al.  Transcriptional dysregulation in Huntington's disease: The role of histone deacetylases. , 2015, Pharmacological research.

[60]  R. Cole,et al.  Phosphorylation of Mutant Huntingtin at Serine 116 Modulates Neuronal Toxicity , 2014, PloS one.

[61]  Pornpimol Charoentong,et al.  ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks , 2009, Bioinform..

[62]  Jaak Vilo,et al.  ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap , 2015, Nucleic Acids Res..

[63]  J. M. Boutell,et al.  Aberrant interactions of transcriptional repressor proteins with the Huntington's disease gene product, huntingtin. , 1999, Human molecular genetics.

[64]  Amanda J. Guise,et al.  Aurora B-dependent Regulation of Class IIa Histone Deacetylases by Mitotic Nuclear Localization Signal Phosphorylation* , 2012, Molecular & Cellular Proteomics.

[65]  C. Ross,et al.  Post-translational modifications clustering within proteolytic domains decrease mutant huntingtin toxicity , 2017, The Journal of Biological Chemistry.

[66]  M. Hayden,et al.  Huntington disease , 2015, Nature Reviews Disease Primers.

[67]  Amanda J. Guise,et al.  Determining the Composition and Stability of Protein Complexes Using an Integrated Label-Free and Stable Isotope Labeling Strategy. , 2016, Methods in molecular biology.

[68]  F. Dequiedt,et al.  Class II histone deacetylases: versatile regulators. , 2003, Trends in genetics : TIG.

[69]  Nan Wang,et al.  Neuronal targets for reducing mutant huntingtin expression to ameliorate disease in a mouse model of Huntington's disease , 2014, Nature Medicine.

[70]  L. DesGroseillers,et al.  The zinc‐finger protein ZFR is critical for Staufen 2 isoform specific nucleocytoplasmic shuttling in neurons , 2006, Journal of neurochemistry.

[71]  Johannes Griss,et al.  The Proteomics Identifications (PRIDE) database and associated tools: status in 2013 , 2012, Nucleic Acids Res..

[72]  D. Dorsett Roles of the sister chromatid cohesion apparatus in gene expression, development, and human syndromes , 2007, Chromosoma.

[73]  W. Duan,et al.  Metabolism in HD: Still a relevant mechanism? , 2014, Movement disorders : official journal of the Movement Disorder Society.

[74]  H. Bading,et al.  Neuronal activity‐dependent nucleocytoplasmic shuttling of HDAC4 and HDAC5 , 2003, Journal of neurochemistry.

[75]  Amy-Joan L Ham,et al.  Sample preparation and digestion for proteomic analyses using spin filters , 2005, Proteomics.

[76]  M. Mann,et al.  Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. , 2003, Analytical chemistry.

[77]  E. Kremmer,et al.  A Novel Sorting Nexin Modulates Endocytic Trafficking and α-Secretase Cleavage of the Amyloid Precursor Protein* , 2008, Journal of Biological Chemistry.

[78]  I. Krantz,et al.  Cornelia de Lange syndrome is caused by mutations in NIPBL, the human homolog of Drosophila melanogaster Nipped-B , 2004, Nature Genetics.