Shared brain transcriptomic signature in TDP-43 type A FTLD patients with or without GRN mutations

Abstract Frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) is a complex heterogeneous neurodegenerative disorder for which mechanisms are poorly understood. To explore transcriptional changes underlying FTLD-TDP, we performed RNA-sequencing on 66 genetically unexplained FTLD-TDP patients, 24 FTLD-TDP patients with GRN mutations and 24 control participants. Using principal component analysis, hierarchical clustering, differential expression and coexpression network analyses, we showed that GRN mutation carriers and FTLD-TDP-A patients without a known mutation shared a common transcriptional signature that is independent of GRN loss-of-function. After combining both groups, differential expression as compared to the control group and coexpression analyses revealed alteration of processes related to immune response, synaptic transmission, RNA metabolism, angiogenesis and vesicle-mediated transport. Deconvolution of the data highlighted strong cellular alterations that were similar in FTLD-TDP-A and GRN mutation carriers with NSF as a potentially important player in both groups. We propose several potentially druggable pathways such as the GABAergic, GDNF and sphingolipid pathways. Our findings underline new disease mechanisms and strongly suggest that affected pathways in GRN mutation carriers extend beyond GRN and contribute to genetically unexplained forms of FTLD-TDP-A.

[1]  Anna L. Brown,et al.  Common ALS/FTD risk variants in UNC13A exacerbate its cryptic splicing and loss upon TDP-43 mislocalization , 2021, bioRxiv.

[2]  Caitlin M. Rodriguez,et al.  TDP-43 represses cryptic exon inclusion in the FTD–ALS gene UNC13A , 2021, Nature.

[3]  K. Herrup,et al.  Myelin pathology in ataxia-telangiectasia is the cell-intrinsic consequence of ATM deficiency in the oligodendrocytes , 2021, medRxiv.

[4]  E. McDonagh,et al.  Open Targets Platform: supporting systematic drug–target identification and prioritisation , 2020, Nucleic Acids Res..

[5]  M. Martinez-Fierro,et al.  The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases , 2020, International journal of molecular sciences.

[6]  T. Carpenter,et al.  GABA and glutamate deficits from frontotemporal lobar degeneration are associated with disinhibition , 2020, Brain : a journal of neurology.

[7]  H. Cao,et al.  Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer’s disease , 2020, Proceedings of the National Academy of Sciences.

[8]  Michelle K. Cahill,et al.  Neurotoxic microglia promote TDP-43 proteinopathy in progranulin deficiency , 2020, Nature.

[9]  Anna L. Brown,et al.  Truncated stathmin-2 is a marker of TDP-43 pathology in frontotemporal dementia. , 2020, The Journal of clinical investigation.

[10]  Y. Asmann,et al.  Loss of TMEM106B leads to myelination deficits: implications for frontotemporal dementia treatment strategies. , 2020, Brain : a journal of neurology.

[11]  R. Vandenberghe,et al.  Distinct molecular patterns of TDP-43 pathology in Alzheimer’s disease: relationship with clinical phenotypes , 2020, Acta Neuropathologica Communications.

[12]  Y. Asmann,et al.  Deciphering cellular transcriptional alterations in Alzheimer’s disease brains , 2020, bioRxiv.

[13]  M. Humphries,et al.  KANK2 Links αVβ5 Focal Adhesions to Microtubules and Regulates Sensitivity to Microtubule Poisons and Cell Migration , 2020, Frontiers in Cell and Developmental Biology.

[14]  L. Petrucelli,et al.  Extensive transcriptomic study emphasizes importance of vesicular transport in C9orf72 expansion carriers , 2019, Acta Neuropathologica Communications.

[15]  B. Brüne,et al.  Sphingosine-1-Phosphate and Macrophage Biology—How the Sphinx Tames the Big Eater , 2019, Front. Immunol..

[16]  E. Englund,et al.  A cortical microvascular structure in vascular dementia, Alzheimer's disease, frontotemporal lobar degeneration and nondemented controls: a sign of angiogenesis due to brain ischaemia? , 2019, Neuropathology and applied neurobiology.

[17]  Olga Tanaseichuk,et al.  Metascape provides a biologist-oriented resource for the analysis of systems-level datasets , 2019, Nature Communications.

[18]  Artur Lugmayr,et al.  CirGO: an alternative circular way of visualising gene ontology terms , 2019, BMC Bioinformatics.

[19]  Kevin F. Bieniek,et al.  Genome-wide analyses as part of the international FTLD-TDP whole-genome sequencing consortium reveals novel disease risk factors and increases support for immune dysfunction in FTLD , 2019, Acta Neuropathologica.

[20]  Michael J. Steinbaugh,et al.  ALS IMPLICATED PROTEIN TDP-43 SUSTAINS LEVELS OF STMN2 A MEDIATOR OF MOTOR NEURON GROWTH AND REPAIR , 2019, Nature Neuroscience.

[21]  F. Rigo,et al.  Premature polyadenylation-mediated loss of stathmin-2 is a hallmark of TDP-43-dependent neurodegeneration , 2018, Nature Neuroscience.

[22]  C. Jack,et al.  Pathological, imaging and genetic characteristics support the existence of distinct TDP-43 types in non-FTLD brains , 2019, Acta Neuropathologica.

[23]  Rajendra K. Sharma,et al.  Calmodulin-binding proteins: A journey of 40 years. , 2018, Cell calcium.

[24]  K. Nave,et al.  Cell-autonomous requirement of TDP-43, an ALS/FTD signature protein, for oligodendrocyte survival and myelination , 2018, Proceedings of the National Academy of Sciences.

[25]  Ying Sun,et al.  Progranulin associates with hexosaminidase A and ameliorates GM2 ganglioside accumulation and lysosomal storage in Tay-Sachs disease , 2018, Journal of Molecular Medicine.

[26]  R. Faull,et al.  Gamma-aminobutyric acid A receptors in Alzheimer's disease: highly localized remodeling of a complex and diverse signaling pathway , 2018, Neural regeneration research.

[27]  R. Sandhoff,et al.  Emerging concepts of ganglioside metabolism , 2018, FEBS letters.

[28]  Panos Roussos,et al.  Brain Cell Type Specific Gene Expression and Co-expression Network Architectures , 2018, Scientific Reports.

[29]  Kevin F. Bieniek,et al.  Potential genetic modifiers of disease risk and age at onset in patients with frontotemporal lobar degeneration and GRN mutations: a genome-wide association study , 2018, Lancet Neurology.

[30]  S. Leurgans,et al.  Frontotemporal dysregulation of the SNARE protein interactome is associated with faster cognitive decline in old age , 2018, Neurobiology of Disease.

[31]  J. Ji,et al.  S1PR3 is essential for phosphorylated fingolimod to protect astrocytes against oxygen‐glucose deprivation‐induced neuroinflammation via inhibiting TLR2/4‐NFκB signalling , 2018, Journal of cellular and molecular medicine.

[32]  R. Proia,et al.  Sphingosine-1-Phosphate and the S1P3 Receptor Initiate Neuronal Retraction via RhoA/ROCK Associated with CRMP2 Phosphorylation , 2017, Front. Mol. Neurosci..

[33]  W. Seeley,et al.  Impaired prosaposin lysosomal trafficking in frontotemporal lobar degeneration due to progranulin mutations , 2017, Nature Communications.

[34]  G. Halliday,et al.  Gene therapy for Parkinson's disease: Disease modification by GDNF family of ligands , 2017, Neurobiology of Disease.

[35]  J. Trojanowski,et al.  Expansion of the classification of FTLD-TDP: distinct pathology associated with rapidly progressive frontotemporal degeneration , 2017, Acta Neuropathologica.

[36]  S. Leurgans,et al.  Presynaptic proteins complexin-I and complexin-II differentially influence cognitive function in early and late stages of Alzheimer’s disease , 2017, Acta Neuropathologica.

[37]  Ying Sun,et al.  Progranulin Recruits HSP70 to β-Glucocerebrosidase and Is Therapeutic Against Gaucher Disease , 2016, EBioMedicine.

[38]  S. Rollinson,et al.  Identification of biological pathways regulated by PGRN and GRN peptide treatments using transcriptome analysis , 2016, The European journal of neuroscience.

[39]  Tanya M. Teslovich,et al.  Prosaposin is a regulator of progranulin levels and oligomerization , 2016, Nature Communications.

[40]  Michelle K. Cahill,et al.  Progranulin Deficiency Promotes Circuit-Specific Synaptic Pruning by Microglia via Complement Activation , 2016, Cell.

[41]  Eric Karran,et al.  The Cellular Phase of Alzheimer’s Disease , 2016, Cell.

[42]  S. Rombouts,et al.  Cerebral blood flow in presymptomatic MAPT and GRN mutation carriers: A longitudinal arterial spin labeling study☆ , 2016, NeuroImage: Clinical.

[43]  N. Belluardo,et al.  Current disease modifying approaches to treat Parkinson’s disease , 2016, Cellular and Molecular Life Sciences.

[44]  L. Petrucelli,et al.  TAR DNA‐binding protein 43 and pathological subtype of Alzheimer's disease impact clinical features , 2015, Annals of neurology.

[45]  Ying Sun,et al.  Prosaposin facilitates sortilin-independent lysosomal trafficking of progranulin , 2015, The Journal of cell biology.

[46]  L. Reid,et al.  Motor pathway degeneration in young ataxia telangiectasia patients: A diffusion tractography study , 2015, NeuroImage: Clinical.

[47]  M. Sperandio,et al.  Sphingosine-1-phosphate receptor 3 promotes leukocyte rolling by mobilizing endothelial P-selectin , 2015, Nature Communications.

[48]  P. Byrd,et al.  Ataxia telangiectasia: more variation at clinical and cellular levels , 2015, Clinical genetics.

[49]  T. Kubo,et al.  Knock Out of S1P3 Receptor Signaling Attenuates Inflammation and Fibrosis in Bleomycin-Induced Lung Injury Mice Model , 2014, PloS one.

[50]  Mike P. Wattjes,et al.  Distinct perfusion patterns in Alzheimer’s disease, frontotemporal dementia and dementia with Lewy bodies , 2014, European Radiology.

[51]  Alexander Gerhard,et al.  Frontotemporal dementia and its subtypes: a genome-wide association study , 2014, The Lancet Neurology.

[52]  Hee-Sun Kim,et al.  The anti-inflammatory role of tissue inhibitor of metalloproteinase-2 in lipopolysaccharide-stimulated microglia , 2014, Journal of Neuroinflammation.

[53]  H. Wilms,et al.  Glial Cell Line-Derived Neurotrophic Factor Family Members Reduce Microglial Activation via Inhibiting p38MAPKs-Mediated Inflammatory Responses , 2014, Journal of neurodegenerative diseases.

[54]  R. Petersen,et al.  Progranulin protein levels are differently regulated in plasma and CSF , 2014, Neurology.

[55]  K. Sandhoff,et al.  Sphingolipids and lysosomal pathologies. , 2014, Biochimica et biophysica acta.

[56]  C. Jack,et al.  TDP-43 is a key player in the clinical features associated with Alzheimer’s disease , 2014, Acta Neuropathologica.

[57]  R. Bartus,et al.  Parkinson's Disease Gene Therapy: Success by Design Meets Failure by Efficacy , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[58]  Andreas Krämer,et al.  Causal analysis approaches in Ingenuity Pathway Analysis , 2013, Bioinform..

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

[60]  H. Voss,et al.  Progranulin Deficiency Promotes Post-Ischemic Blood–Brain Barrier Disruption , 2013, The Journal of Neuroscience.

[61]  D. Cleveland,et al.  Converging Mechanisms in ALS and FTD: Disrupted RNA and Protein Homeostasis , 2013, Neuron.

[62]  L. S. Sefcik,et al.  Sphingosine 1-phosphate receptor 3 regulates recruitment of anti-inflammatory monocytes to microvessels during implant arteriogenesis , 2013, Proceedings of the National Academy of Sciences.

[63]  A. Bateman,et al.  Expression of the Growth Factor Progranulin in Endothelial Cells Influences Growth and Development of Blood Vessels: A Novel Mouse Model , 2013, PloS one.

[64]  James B. Rowe,et al.  Reorganisation of brain networks in frontotemporal dementia and progressive supranuclear palsy☆ , 2013, NeuroImage: Clinical.

[65]  J. Chun,et al.  Lysophospholipids and their receptors in the central nervous system. , 2013, Biochimica et biophysica acta.

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

[67]  Yi Zhong,et al.  Digital sorting of complex tissues for cell type-specific gene expression profiles , 2013, BMC Bioinformatics.

[68]  Wei Li,et al.  RSeQC: quality control of RNA-seq experiments , 2012, Bioinform..

[69]  Davis J. McCarthy,et al.  Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation , 2012, Nucleic acids research.

[70]  R. Faull,et al.  Population-specific expression analysis (PSEA) reveals molecular changes in diseased brain , 2011, Nature Methods.

[71]  Bruce L. Miller,et al.  Expanded GGGGCC Hexanucleotide Repeat in Noncoding Region of C9ORF72 Causes Chromosome 9p-Linked FTD and ALS , 2011, Neuron.

[72]  David Heckerman,et al.  A Hexanucleotide Repeat Expansion in C9ORF72 Is the Cause of Chromosome 9p21-Linked ALS-FTD , 2011, Neuron.

[73]  Matko Bosnjak,et al.  REVIGO Summarizes and Visualizes Long Lists of Gene Ontology Terms , 2011, PloS one.

[74]  J. Trojanowski,et al.  A harmonized classification system for FTLD-TDP pathology , 2011, Acta Neuropathologica.

[75]  H. Feldman,et al.  rs5848 polymorphism and serum progranulin level , 2011, Journal of the Neurological Sciences.

[76]  D. Selkoe Alzheimer's disease. , 2011, Cold Spring Harbor perspectives in biology.

[77]  Longxuan Li,et al.  Microglial activation state exerts a biphasic influence on brain endothelial cell proliferation by regulating the balance of TNF and TGF-β1 , 2010, Journal of Neuroinflammation.

[78]  J. Trojanowski,et al.  Distinct cerebral perfusion patterns in FTLD and AD , 2010, Neurology.

[79]  G. Bing,et al.  Glial cell line-derived neurotrophic factor protects midbrain dopaminergic neurons against lipopolysaccharide neurotoxicity , 2010, Journal of Neuroimmunology.

[80]  M. J. Fresnadillo Martínez,et al.  Common variants at 7p21 are associated with frontotemporal lobar degeneration with TDP-43 inclusions , 2010, Nature Genetics.

[81]  Norbert Schuff,et al.  Concordance and Discordance Between Brain Perfusion and Atrophy in Frontotemporal Dementia , 2010, Brain Imaging and Behavior.

[82]  J. Trojanowski,et al.  Brain progranulin expression in GRN-associated frontotemporal lobar degeneration , 2009, Acta Neuropathologica.

[83]  Steve Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[84]  R. Petersen,et al.  Common variation in the miR-659 binding-site of GRN is a major risk factor for TDP43-positive frontotemporal dementia , 2008, Human molecular genetics.

[85]  T. Kohriyama,et al.  White matter lesions in the brain with frontotemporal lobar degeneration with motor neuron disease: TDP-43-immunopositive inclusions co-localize with p62, but not ubiquitin , 2008, Acta Neuropathologica.

[86]  J. Trojanowski,et al.  Variations in the progranulin gene affect global gene expression in frontotemporal lobar degeneration. , 2008, Human molecular genetics.

[87]  C. Saper,et al.  Prostaglandin E2 Attenuates Preoptic Expression of GABAA Receptors via EP3 Receptors* , 2008, Journal of Biological Chemistry.

[88]  M. Lavin,et al.  ATM Activation and DNA Damage Response , 2007, Cell cycle.

[89]  Murray Grossman,et al.  TDP-43-Positive White Matter Pathology in Frontotemporal Lobar Degeneration With Ubiquitin-Positive Inclusions , 2007, Journal of neuropathology and experimental neurology.

[90]  G. van Echten-Deckert,et al.  Sphingolipid metabolism in neural cells. , 2006, Biochimica et biophysica acta.

[91]  H. Feldman,et al.  The neuropathology of frontotemporal lobar degeneration caused by mutations in the progranulin gene. , 2006, Brain : a journal of neurology.

[92]  N. Schuff,et al.  Hypoperfusion in frontotemporal dementia and Alzheimer disease by arterial spin labeling MRI , 2006, Neurology.

[93]  C. Duijn,et al.  Null mutations in progranulin cause ubiquitin-positive frontotemporal dementia linked to chromosome 17q21 , 2006, Nature.

[94]  S. Melquist,et al.  Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17 , 2006, Nature.

[95]  S. Tzeng,et al.  Regulation of microglial activities by glial cell line derived neurotrophic factor , 2006, Journal of cellular biochemistry.

[96]  Yosef Shiloh,et al.  Ataxia-telangiectasia and the ATM gene: Linking neurodegeneration, immunodeficiency, and cancer to cell cycle checkpoints , 1996, Journal of Clinical Immunology.

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

[98]  I. Goldberg,et al.  Complex roles of tissue inhibitors of metalloproteinases in cancer , 2002, Oncogene.

[99]  T. Südhof,et al.  Three-Dimensional Structure of the Complexin/SNARE Complex , 2002, Neuron.

[100]  J. Gu,et al.  Integrin α1β1-Mediated Activation of Cyclin-Dependent Kinase 5 Activity Is Involved in Neurite Outgrowth and Human Neurofilament Protein H Lys-Ser-Pro Tail Domain Phosphorylation , 2000, The Journal of Neuroscience.

[101]  K. Khanna Cancer risk and the ATM gene: a continuing debate. , 2000, Journal of the National Cancer Institute.

[102]  I. Ferrer Neurons and Their Dendrites in Frontotemporal Dementia , 1999, Dementia and Geriatric Cognitive Disorders.

[103]  Ronald C. Petersen,et al.  Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17 , 1998, Nature.

[104]  D. Toriumi,et al.  Peripheral nerve regeneration: comparison of laminin and acidic fibroblast growth factor. , 1998, American journal of otolaryngology.

[105]  C. Deng,et al.  Atm selectively regulates distinct p53-dependent cell-cycle checkpoint and apoptotic pathways , 1997, Nature Genetics.

[106]  Thomas C. Südhof,et al.  Complexins: Cytosolic proteins that regulate SNAP receptor function , 1995, Cell.

[107]  Paul Tempst,et al.  SNAP receptors implicated in vesicle targeting and fusion , 1993, Nature.

[108]  Richard O. Hynes,et al.  Integrins: Versatility, modulation, and signaling in cell adhesion , 1992, Cell.

[109]  J. Rothman,et al.  SNAPs, a family of NSF attachment proteins involved in intracellular membrane fusion in animals and yeast , 1990, Cell.

[110]  M. Hemler VLA proteins in the integrin family: structures, functions, and their role on leukocytes. , 1990, Annual review of immunology.

[111]  B. M. Klichev [Conservative treatment of rectal prolapse in children]. , 1989, Pediatriia.

[112]  K. Reid,et al.  Subunit composition and structure of subcomponent C1q of the first component of human complement. , 1976, The Biochemical journal.