RNA-binding deficient TDP-43 drives cognitive decline in a mouse model of TDP-43 proteinopathy
暂无分享,去创建一个
Viktoriya D. Nikolova | S. Moy | Jeremy M. Simon | J. A. Ezzell | Ariana B. Marquez | Deepa Ajit | Julie C. Necarsulmer | Jeremy Simon | Baggio Evangelista | Youjun Chen | Xu Tian | Sara Nafees | Ariana Marquez Gonzalez | Ping Wang | Viktoriya Nikolova | Kathryn Harper | Jennifer Ezzell | Adriana Beltran | Sheryl Moy | Todd Cohen | Huijun Jiang | Feng-Chang Lin | Baggio A. Evangelista | Xu Tian | Kathryn M Harper | Adriana S Beltran | B. A. Evangelista | Todd J Cohen
[1] Adekunle T. Bademosi,et al. Aggregation-prone TDP-43 sequesters and drives pathological transitions of free nuclear TDP-43 , 2023, Cellular and Molecular Life Sciences.
[2] Tracy J. Yuen,et al. Disease-associated oligodendrocyte responses across neurodegenerative diseases. , 2022, Cell reports.
[3] J. V. van Swieten,et al. Neurovascular dysfunction in GRN-associated frontotemporal dementia identified by single-nucleus RNA sequencing of human cerebral cortex , 2022, Nature Neuroscience.
[4] L. Schurgers,et al. Ageing - Oxidative stress, PTMs and disease. , 2022, Molecular aspects of medicine.
[5] Alexander J. Bryer,et al. Recognition of the TDP-43 nuclear localization signal by importin α1/β , 2022, Cell reports.
[6] Marius Pachitariu,et al. Cellpose 2.0: how to train your own model , 2022, bioRxiv.
[7] Sterling C. Johnson,et al. Exploring common genetic contributors to neuroprotection from amyloid pathology , 2022, Brain communications.
[8] E. Buratti,et al. Sirtuin-1 sensitive lysine-136 acetylation drives phase separation and pathological aggregation of TDP-43 , 2022, Nature Communications.
[9] Anna L. Brown,et al. TDP-43 loss and ALS-risk SNPs drive mis-splicing and depletion of UNC13A , 2022, Nature.
[10] D. Dickson,et al. TDP-43 Pathology in Alzheimer’s Disease , 2021, Molecular neurodegeneration.
[11] D. Dickson,et al. TDP-43 Pathology in Alzheimer’s Disease , 2021, Molecular Neurodegeneration.
[12] T. Miller,et al. Specific RNA interactions promote TDP‐43 multivalent phase separation and maintain liquid properties , 2021, EMBO reports.
[13] Yaoyang Zhang,et al. Loss of TDP-43 function underlies hippocampal and cortical synaptic deficits in TDP-43 proteinopathies , 2021, Molecular Psychiatry.
[14] T. Raj,et al. Transcriptomic analysis of frontotemporal lobar degeneration with TDP-43 pathology reveals cellular alterations across multiple brain regions , 2021, Acta Neuropathologica.
[15] S. Bardien,et al. Emerging evidence implicating a role for neurexins in neurodegenerative and neuropsychiatric disorders , 2021, Open Biology.
[16] J. Rothstein,et al. Nuclear RNA binding regulates TDP-43 nuclear localization and passive nuclear export , 2021, bioRxiv.
[17] Nicolas L. Fawzi,et al. TDP-43 condensation properties specify its RNA-binding and regulatory repertoire , 2021, Cell.
[18] D. Dormann,et al. Post-translational modifications on RNA-binding proteins: accelerators, brakes, or passengers in neurodegeneration? , 2021, Trends in biochemical sciences.
[19] M. Kipp,et al. What Guides Peripheral Immune Cells into the Central Nervous System? , 2021, Cells.
[20] J. Mann,et al. RNA modulates physiological and neuropathological protein phase transitions , 2021, Neuron.
[21] Anne E Carpenter,et al. CellProfiler 4: improvements in speed, utility and usability , 2021, BMC Bioinformatics.
[22] Alvaro A. Beltran,et al. Generation of an induced pluripotent stem cell line (UNCCi002-A) from a healthy donor using a non-integration system to study Cerebral Cavernous Malformation (CCM). , 2021, Stem cell research.
[23] Nicole F. Liachko,et al. Regulation of TDP-43 phosphorylation in aging and disease , 2021, GeroScience.
[24] Xianzheng Sha,et al. Comparative Analysis of Multiple Neurodegenerative Diseases Based on Advanced Epigenetic Aging Brain , 2021, Frontiers in Genetics.
[25] Robert H. Brown,et al. Low-level overexpression of wild type TDP-43 causes late-onset, progressive neurodegeneration and paralysis in mice , 2021, bioRxiv.
[26] G. Hummer,et al. Disease‐linked TDP‐43 hyperphosphorylation suppresses TDP‐43 condensation and aggregation , 2021, bioRxiv.
[27] D. Rincon-Limas,et al. Molecular, functional, and pathological aspects of TDP-43 fragmentation , 2021, iScience.
[28] Caitlin M. Rodriguez,et al. TDP-43 represses cryptic exon inclusion in the FTD–ALS gene UNC13A , 2021, Nature.
[29] J. Yates,et al. HSP70 chaperones RNA-free TDP-43 into anisotropic intranuclear liquid spherical shells , 2020, Science.
[30] P. van Damme,et al. TDP-43 proteinopathies: a new wave of neurodegenerative diseases , 2020, Journal of Neurology, Neurosurgery, and Psychiatry.
[31] Hanna S. Yuan,et al. Frontotemporal dementia‐linked P112H mutation of TDP‐43 induces protein structural change and impairs its RNA binding function , 2020, Protein science : a publication of the Protein Society.
[32] Zuguang Gu,et al. simplifyEnrichment: A Bioconductor Package for Clustering and Visualizing Functional Enrichment Results , 2020, bioRxiv.
[33] D. E. López,et al. Prepulse Inhibition of the Auditory Startle Reflex Assessment as a Hallmark of Brainstem Sensorimotor Gating Mechanisms , 2020, Brain sciences.
[34] Anna L. Brown,et al. Truncated stathmin-2 is a marker of TDP-43 pathology in frontotemporal dementia. , 2020, The Journal of clinical investigation.
[35] Jie Zhang,et al. Attenuation of epigenetic regulator SMARCA4 and ERK‐ETS signaling suppresses aging‐related dopaminergic degeneration , 2020, Aging cell.
[36] T. Shimamura,et al. ARHGAP10, which encodes Rho GTPase-activating protein 10, is a novel gene for schizophrenia risk , 2020, Translational Psychiatry.
[37] Robert Johnson,et al. Structural Analysis , 2020, Multiphysics Modeling with Application to Biomedical Engineering.
[38] J. Vilo,et al. gprofiler2 -- an R package for gene list functional enrichment analysis and namespace conversion toolset g:Profiler , 2020, F1000Research.
[39] R. Vandenberghe,et al. Distinct molecular patterns of TDP-43 pathology in Alzheimer’s disease: relationship with clinical phenotypes , 2020, Acta Neuropathologica Communications.
[40] Samuel J. Lord,et al. SuperPlots: Communicating reproducibility and variability in cell biology , 2020, The Journal of cell biology.
[41] Marius Pachitariu,et al. Cellpose: a generalist algorithm for cellular segmentation , 2020, Nature Methods.
[42] S. Mirarab,et al. Sequence Analysis , 2020, Encyclopedia of Bioinformatics and Computational Biology.
[43] E. Tseng,et al. 5′UTR-mediated regulation of Ataxin-1 expression , 2020, Neurobiology of Disease.
[44] Pei-Lin Cheng,et al. A robust TDP-43 knock-in mouse model of ALS , 2020, Acta Neuropathologica Communications.
[45] M. Khanna,et al. Structural Insights Into TDP-43 and Effects of Post-translational Modifications , 2019, Front. Mol. Neurosci..
[46] Lilah M. Besser,et al. Limbic Predominant Age-Related TDP-43 Encephalopathy (LATE): Clinical and Neuropathological Associations. , 2019, Journal of neuropathology and experimental neurology.
[47] R. Irizarry. ggplot2 , 2019, Introduction to Data Science.
[48] Jie Zhang,et al. Attenuation of epigenetic regulator SMARCA4 and ERK-ETS signaling suppresses aging-related dopaminergic degeneration , 2019, bioRxiv.
[49] Samuel J. Lord,et al. If your P value looks too good to be true, it probably is: Communicating reproducibility and variability in cell biology , 2019, 1911.03509.
[50] Laura E. Herring,et al. Site-specific phosphorylation and caspase cleavage of GFAP are new markers of Alexander disease severity , 2019, eLife.
[51] R. Rohatgi,et al. Phase separation-deficient TDP43 remains functional in splicing , 2019, Nature Communications.
[52] C. Shaw,et al. RRM adjacent TARDBP mutations disrupt RNA binding and enhance TDP-43 proteinopathy , 2019, Brain : a journal of neurology.
[53] M. Hasegawa,et al. The basis of clinicopathological heterogeneity in TDP-43 proteinopathy , 2019, Acta Neuropathologica.
[54] R. Hertzano,et al. Semaphorin-5B Controls Spiral Ganglion Neuron Branch Refinement during Development , 2019, The Journal of Neuroscience.
[55] J. Rohrer,et al. An update on genetic frontotemporal dementia , 2019, Journal of Neurology.
[56] J. Vilo,et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update) , 2019, Nucleic Acids Res..
[57] C. Jack,et al. Limbic-predominant age-related TDP-43 encephalopathy (LATE): consensus working group report , 2019, Brain : a journal of neurology.
[58] A. Walker,et al. The Pathobiology of TDP-43 C-Terminal Fragments in ALS and FTLD , 2019, Front. Neurosci..
[59] Lin Guo,et al. Cytoplasmic TDP-43 De-mixing Independent of Stress Granules Drives Inhibition of Nuclear Import, Loss of Nuclear TDP-43, and Cell Death , 2019, Neuron.
[60] B. Portz,et al. RNA Binding Antagonizes Neurotoxic Phase Transitions of TDP-43 , 2019, Neuron.
[61] Trees-Juen Chuang,et al. Transcriptomopathies of pre- and post-symptomatic frontotemporal dementia-like mice with TDP-43 depletion in forebrain neurons , 2019, Acta Neuropathologica Communications.
[62] Qi Zhang,et al. Regional and Cellular Mapping of Sortilin Immunoreactivity in Adult Human Brain , 2019, Front. Neuroanat..
[63] R. Rohatgi,et al. Decoding and recoding phase behavior of TDP43 reveals that phase separation is not required for splicing function , 2019, bioRxiv.
[64] C. Ibáñez,et al. Abnormal TDP‐43 function impairs activity‐dependent BDNF secretion, synaptic plasticity, and cognitive behavior through altered Sortilin splicing , 2019, The EMBO journal.
[65] N. Shneider,et al. Mutant TDP-43 Causes Early-Stage Dose-Dependent Motor Neuron Degeneration in a TARDBP Knockin Mouse Model of ALS. , 2019, Cell reports.
[66] F. Rigo,et al. Premature polyadenylation-mediated loss of stathmin-2 is a hallmark of TDP-43-dependent neurodegeneration , 2018, Nature Neuroscience.
[67] Shinji Ogaki,et al. Arnold , 2018, ACM Trans. Graph..
[68] Matthew A. White,et al. Publisher Correction: TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD , 2018, Nature Neuroscience.
[69] P. Tomançak,et al. RNA buffers the phase separation behavior of prion-like RNA binding proteins , 2018, Science.
[70] Jeremy Stinson,et al. CRISPR off-target analysis in genetically engineered rats and mice , 2018, Nature Methods.
[71] Eric T. Wang,et al. Mice with endogenous TDP‐43 mutations exhibit gain of splicing function and characteristics of amyotrophic lateral sclerosis , 2018, The EMBO journal.
[72] Maximilian Haeussler,et al. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens , 2018, Nucleic Acids Res..
[73] Y. Chook,et al. Active nuclear import and passive nuclear export are the primary determinants of TDP-43 localization , 2018, Scientific Reports.
[74] W. Ge,et al. Roles of Nitric Oxide Synthase Isoforms in Neurogenesis , 2018, Molecular Neurobiology.
[75] Luwen Wang,et al. Pathomechanisms of TDP‐43 in neurodegeneration , 2018, Journal of neurochemistry.
[76] Matthew A. White,et al. TDP-43 gains function due to perturbed autoregulation in a Tardbp knock-in mouse model of ALS-FTD , 2018, Nature Neuroscience.
[77] E. Buratti. TDP-43 post-translational modifications in health and disease , 2018, Expert opinion on therapeutic targets.
[78] David A. Knowles,et al. Annotation-free quantification of RNA splicing using LeafCutter , 2017, Nature Genetics.
[79] Michael S. Bereman,et al. Acetylation-induced TDP-43 pathology is suppressed by an HSF1-dependent chaperone program , 2017, Nature Communications.
[80] E. Masliah,et al. Importance of adiponectin activity in the pathogenesis of Alzheimer's disease , 2017, Annals of clinical and translational neurology.
[81] Hadley Wickham,et al. ggplot2 - Elegant Graphics for Data Analysis (2nd Edition) , 2017 .
[82] Alexander Lex,et al. UpSetR: an R package for the visualization of intersecting sets and their properties , 2017, bioRxiv.
[83] Geet Duggal,et al. Salmon: fast and bias-aware quantification of transcript expression using dual-phase inference , 2017, Nature Methods.
[84] Marco Prinz,et al. The role of peripheral immune cells in the CNS in steady state and disease , 2017, Nature Neuroscience.
[85] P. Wong,et al. Depletion of TDP-43 decreases fibril and plaque β-amyloid and exacerbates neurodegeneration in an Alzheimer’s mouse model , 2016, Acta Neuropathologica.
[86] L. G. De la Casa,et al. Reduced Prepulse Inhibition as a Biomarker of Schizophrenia , 2016, Front. Behav. Neurosci..
[87] W. Scheper,et al. Targeting neuronal MAPK14/p38α activity to modulate autophagy in the Alzheimer disease brain , 2016, Autophagy.
[88] V. Plagnol,et al. Quantitative analysis of cryptic splicing associated with TDP-43 depletion , 2016, BMC Medical Genomics.
[89] R. Eils,et al. Complex heatmaps reveal patterns and correlations in multidimensional genomic data , 2016, Bioinform..
[90] J. Gallo,et al. Tau mis-splicing in the pathogenesis of neurodegenerative disorders , 2016, BMB reports.
[91] J. Hodges,et al. The frontotemporal dementia-motor neuron disease continuum , 2016, The Lancet.
[92] C. Dobson,et al. Quantification of the Relative Contributions of Loss-of-function and Gain-of-function Mechanisms in TAR DNA-binding Protein 43 (TDP-43) Proteinopathies * , 2016, The Journal of Biological Chemistry.
[93] J. Joly,et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR , 2016, Genome Biology.
[94] I. Mackenzie,et al. Molecular neuropathology of frontotemporal dementia: insights into disease mechanisms from postmortem studies , 2016, Journal of neurochemistry.
[95] Eric E. Smith,et al. The Prevalence and Incidence of Frontotemporal Dementia: a Systematic Review , 2016, Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques.
[96] D. Bennett,et al. Therapeutic correction of ApoER2 splicing in Alzheimer's disease mice using antisense oligonucleotides , 2016, EMBO molecular medicine.
[97] C. Lim,et al. Structural analysis of disease-related TDP-43 D169G mutation: linking enhanced stability and caspase cleavage efficiency to protein accumulation , 2016, Scientific Reports.
[98] L. Petrucelli,et al. TDP-43 functions within a network of hnRNP proteins to inhibit the production of a truncated human SORT1 receptor. , 2016, Human molecular genetics.
[99] K. Tenbrock,et al. Neurodegeneration Triggers Peripheral Immune Cell Recruitment into the Forebrain , 2016, The Journal of Neuroscience.
[100] Meagan E. Sullender,et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.
[101] W. Arnold,et al. Electrophysiological Motor Unit Number Estimation (MUNE) Measuring Compound Muscle Action Potential (CMAP) in Mouse Hindlimb Muscles , 2015, Journal of visualized experiments : JoVE.
[102] P. Wong,et al. TDP-43 repression of nonconserved cryptic exons is compromised in ALS-FTD , 2015, Science.
[103] J. Trojanowski,et al. Functional recovery in new mouse models of ALS/FTLD after clearance of pathological cytoplasmic TDP-43 , 2015, Acta Neuropathologica.
[104] F. Cavalcanti,et al. Splicing: is there an alternative contribution to Parkinson’s disease? , 2015, neurogenetics.
[105] M. Rosenfeld,et al. LRP8-Reelin-Regulated Neuronal Enhancer Signature Underlying Learning and Memory Formation , 2015, Neuron.
[106] M. L. Seibenhener,et al. Use of the Open Field Maze to measure locomotor and anxiety-like behavior in mice. , 2015, Journal of visualized experiments : JoVE.
[107] Matthew E. Ritchie,et al. limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.
[108] J. Trojanowski,et al. An acetylation switch controls TDP-43 function and aggregation propensity , 2015, Nature Communications.
[109] A. Kolodkin,et al. Semaphorin 5A inhibits synaptogenesis in early postnatal- and adult-born hippocampal dentate granule cells , 2014, eLife.
[110] T. Willnow,et al. Sorting receptor sortilin—a culprit in cardiovascular and neurological diseases , 2014, Journal of Molecular Medicine.
[111] Robert H. Brown,et al. Partial loss of TDP-43 function causes phenotypes of amyotrophic lateral sclerosis , 2014, Proceedings of the National Academy of Sciences.
[112] H. Fuchs,et al. Mitochondrial Dysfunction and Decrease in Body Weight of a Transgenic Knock-in Mouse Model for TDP-43* , 2014, The Journal of Biological Chemistry.
[113] P. Callaerts,et al. TDP-43-mediated neurodegeneration: towards a loss-of-function hypothesis? , 2014, Trends in molecular medicine.
[114] P. Kuo,et al. The crystal structure of TDP-43 RRM1-DNA complex reveals the specific recognition for UG- and TG-rich nucleic acids , 2014, Nucleic acids research.
[115] J. Ule,et al. Molecular basis of UG-rich RNA recognition by the human splicing factor TDP-43 , 2013, Nature Structural &Molecular Biology.
[116] K. Fox,et al. The role of nitric oxide in pre-synaptic plasticity and homeostasis , 2013, Front. Cell. Neurosci..
[117] W. Wurst,et al. Expression Analysis of Lrrk1, Lrrk2 and Lrrk2 Splice Variants in Mice , 2013, PloS one.
[118] G. Sobue,et al. Loss of TDP-43 causes age-dependent progressive motor neuron degeneration. , 2013, Brain : a journal of neurology.
[119] Gene W. Yeo,et al. ALS-linked TDP-43 mutations produce aberrant RNA splicing and adult-onset motor neuron disease without aggregation or loss of nuclear TDP-43 , 2013, Proceedings of the National Academy of Sciences.
[120] A. Al-Chalabi,et al. Loss and gain of Drosophila TDP-43 impair synaptic efficacy and motor control leading to age-related neurodegeneration by loss-of-function phenotypes , 2013, Human molecular genetics.
[121] L. Petrucelli,et al. Misregulation of human sortilin splicing leads to the generation of a nonfunctional progranulin receptor , 2012, Proceedings of the National Academy of Sciences.
[122] F. J. Livesey,et al. Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks , 2012, Nature Protocols.
[123] C. van Broeckhoven,et al. The genetics and neuropathology of frontotemporal lobar degeneration , 2012, Acta Neuropathologica.
[124] N. Cairns,et al. Mechanisms of disease in frontotemporal lobar degeneration: gain of function versus loss of function effects , 2012, Acta Neuropathologica.
[125] A. Cardona,et al. Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.
[126] Allissa Dillman,et al. MAPT expression and splicing is differentially regulated by brain region: relation to genotype and implication for tauopathies , 2012, Human molecular genetics.
[127] O. Levy,et al. The neuropathology of genetic Parkinson's disease , 2012, Movement disorders : official journal of the Movement Disorder Society.
[128] E. Buratti,et al. TDP-43: gumming up neurons through protein-protein and protein-RNA interactions. , 2012, Trends in biochemical sciences.
[129] K. Manova-Todorova,et al. Tunneling Nanotubes Provide a Unique Conduit for Intercellular Transfer of Cellular Contents in Human Malignant Pleural Mesothelioma , 2012, PloS one.
[130] J. Trojanowski,et al. Redox signalling directly regulates TDP‐43 via cysteine oxidation and disulphide cross‐linking , 2012, The EMBO journal.
[131] C. Vaegter,et al. Sortilin and SorLA Regulate Neuronal Sorting of Trophic and Dementia-Linked Proteins , 2012, Molecular Neurobiology.
[132] E. Buratti,et al. Cellular Model of TAR DNA-binding Protein 43 (TDP-43) Aggregation Based on Its C-terminal Gln/Asn-rich Region* , 2012, The Journal of Biological Chemistry.
[133] J. Trojanowski,et al. Gains or losses: molecular mechanisms of TDP43-mediated neurodegeneration , 2011, Nature Reviews Neuroscience.
[134] Xiaowei Wang,et al. PrimerBank: a PCR primer database for quantitative gene expression analysis, 2012 update , 2011, Nucleic Acids Res..
[135] J. Trojanowski,et al. TDP-43 functions and pathogenic mechanisms implicated in TDP-43 proteinopathies. , 2011, Trends in molecular medicine.
[136] J. Hodges,et al. Motor neuron dysfunction in frontotemporal dementia. , 2011, Brain : a journal of neurology.
[137] B. Tu,et al. Hyperphosphorylation as a Defense Mechanism to Reduce TDP-43 Aggregation , 2011, PloS one.
[138] E. Buratti,et al. TDP-43 Autoregulation: Implications for Disease , 2011, Journal of Molecular Neuroscience.
[139] E. Weeber,et al. Similarities and differences in structure, expression, and functions of VLDLR and ApoER2 , 2011, Molecular Neurodegeneration.
[140] Marcel Martin. Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .
[141] Gene W. Yeo,et al. Long pre-mRNA depletion and RNA missplicing contribute to neuronal vulnerability from loss of TDP-43 , 2011, Nature Neuroscience.
[142] J. Ule,et al. Characterising the RNA targets and position-dependent splicing regulation by TDP-43; implications for neurodegenerative diseases , 2011, Nature Neuroscience.
[143] Michael J. Ziller,et al. Reference Maps of Human ES and iPS Cell Variation Enable High-Throughput Characterization of Pluripotent Cell Lines , 2011, Cell.
[144] J. Trojanowski,et al. Dysregulation of the ALS-associated gene TDP-43 leads to neuronal death and degeneration in mice. , 2011, The Journal of clinical investigation.
[145] A. Naldi,et al. Dementia and cognitive impairment in amyotrophic lateral sclerosis: a review , 2011, Neurological Sciences.
[146] Jernej Ule,et al. TDP‐43 regulates its mRNA levels through a negative feedback loop , 2011, The EMBO journal.
[147] Daniel R. Dries,et al. TDP-43 Is Directed to Stress Granules by Sorbitol, a Novel Physiological Osmotic and Oxidative Stressor , 2010, Molecular and Cellular Biology.
[148] H. Feldman,et al. Sortilin-Mediated Endocytosis Determines Levels of the Frontotemporal Dementia Protein, Progranulin , 2010, Neuron.
[149] H. Hutter,et al. Neurotoxic effects of TDP-43 overexpression in C. elegans. , 2010, Human molecular genetics.
[150] L. Petrucelli,et al. Wild-Type Human TDP-43 Expression Causes TDP-43 Phosphorylation, Mitochondrial Aggregation, Motor Deficits, and Early Mortality in Transgenic Mice , 2010, The Journal of Neuroscience.
[151] Pedro M. Valero-Mora,et al. ggplot2: Elegant Graphics for Data Analysis , 2010 .
[152] E. Buratti,et al. The multiple roles of TDP-43 in pre-mRNA processing and gene expression regulation , 2010, RNA biology.
[153] J. Trojanowski,et al. Amyotrophic lateral sclerosis and frontotemporal lobar degeneration: A spectrum of TDP‐43 proteinopathies , 2010, Neuropathology : official journal of the Japanese Society of Neuropathology.
[154] G. Schellenberg,et al. Loss of murine TDP-43 disrupts motor function and plays an essential role in embryogenesis , 2010, Acta Neuropathologica.
[155] E. Buratti,et al. TDP‐43 is recruited to stress granules in conditions of oxidative insult , 2009, Journal of neurochemistry.
[156] John Q Trojanowski,et al. Mutations in TDP-43 link glycine-rich domain functions to amyotrophic lateral sclerosis. , 2009, Human molecular genetics.
[157] J. Morris,et al. TARDBP 3′-UTR variant in autopsy-confirmed frontotemporal lobar degeneration with TDP-43 proteinopathy , 2009, Acta Neuropathologica.
[158] T. O'Connor,et al. Semaphorin 5B mediates synapse elimination in hippocampal neurons , 2009, Neural Development.
[159] M. Tomishima,et al. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling , 2009, Nature Biotechnology.
[160] John L. Robinson,et al. Clinical and pathological continuum of multisystem TDP-43 proteinopathies. , 2009, Archives of neurology.
[161] J. Trojanowski,et al. Phosphorylation of S409/410 of TDP-43 is a consistent feature in all sporadic and familial forms of TDP-43 proteinopathies , 2009, Acta Neuropathologica.
[162] M. Neumann,et al. Molecular Neuropathology of TDP-43 Proteinopathies , 2009, International journal of molecular sciences.
[163] Andrea D'Ambrogio,et al. Structural determinants of the cellular localization and shuttling of TDP-43 , 2008, Journal of Cell Science.
[164] Christian Büchel,et al. Dissociable Roles for the Hippocampus and the Amygdala in Human Cued versus Context Fear Conditioning , 2008, The Journal of Neuroscience.
[165] J. Trojanowski,et al. Enrichment of C-terminal fragments in TAR DNA-binding protein-43 cytoplasmic inclusions in brain but not in spinal cord of frontotemporal lobar degeneration and amyotrophic lateral sclerosis. , 2008, The American journal of pathology.
[166] J. Trojanowski,et al. Variations in the progranulin gene affect global gene expression in frontotemporal lobar degeneration. , 2008, Human molecular genetics.
[167] Eric R. Kandel,et al. Transgenic Mice Lacking NMDAR-Dependent LTD Exhibit Deficits in Behavioral Flexibility , 2008, Neuron.
[168] Xun Hu,et al. TDP-43 Mutations in Familial and Sporadic Amyotrophic Lateral Sclerosis , 2008, Science.
[169] J. Trojanowski,et al. TDP-43 proteinopathy in frontotemporal lobar degeneration and amyotrophic lateral sclerosis: protein misfolding diseases without amyloidosis. , 2007, Archives of neurology.
[170] D. Butterfield,et al. Nitric oxide in the central nervous system: neuroprotection versus neurotoxicity , 2007, Nature Reviews Neuroscience.
[171] John L. Robinson,et al. Co-morbidity of TDP-43 proteinopathy in Lewy body related diseases , 2007, Acta Neuropathologica.
[172] J. Morris,et al. TDP-43 in familial and sporadic frontotemporal lobar degeneration with ubiquitin inclusions. , 2007, The American journal of pathology.
[173] T. Paunesku,et al. Gene expression analysis of frontotemporal lobar degeneration of the motor neuron disease type with ubiquitinated inclusions , 2007, Acta Neuropathologica.
[174] 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.
[175] M. Jung,et al. Neural circuits and mechanisms involved in Pavlovian fear conditioning: A critical review , 2006, Neuroscience & Biobehavioral Reviews.
[176] Anne E Carpenter,et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes , 2006, Genome Biology.
[177] Bruce L. Miller,et al. Ubiquitinated TDP-43 in Frontotemporal Lobar Degeneration and Amyotrophic Lateral Sclerosis , 2006, Science.
[178] S. Melquist,et al. Mutations in progranulin cause tau-negative frontotemporal dementia linked to chromosome 17 , 2006, Nature.
[179] C. Vorhees,et al. Morris water maze: procedures for assessing spatial and related forms of learning and memory , 2006, Nature Protocols.
[180] N. Sato,et al. Prepulse inhibition of acoustic startle response in mild cognitive impairment and mild dementia of Alzheimer type , 2006, Psychiatry and clinical neurosciences.
[181] M. Frotscher,et al. Modulation of Synaptic Plasticity and Memory by Reelin Involves Differential Splicing of the Lipoprotein Receptor Apoer2 , 2005, Neuron.
[182] F. X. Guix,et al. The physiology and pathophysiology of nitric oxide in the brain , 2005, Progress in Neurobiology.
[183] Michael J. Hansen,et al. Semaphorin 5A Is a Bifunctional Axon Guidance Cue Regulated by Heparan and Chondroitin Sulfate Proteoglycans , 2004, Neuron.
[184] E. Brunamonti,et al. Evaluation of the elevated plus-maze and open-field tests for the assessment of anxiety-related behaviour in inbred mice , 2002, Behavioural Brain Research.
[185] S Shimohama,et al. Activation of MKK6, an upstream activator of p38, in Alzheimer's disease , 2001, Journal of neurochemistry.
[186] Francisco E. Baralle,et al. Characterization and Functional Implications of the RNA Binding Properties of Nuclear Factor TDP-43, a Novel Splicing Regulator ofCFTR Exon 9* , 2001, The Journal of Biological Chemistry.
[187] Jeremy M. Shefner,et al. Motor unit number estimation in human neurological diseases and animal models , 2001, Clinical Neurophysiology.
[188] M. Pfaffl,et al. A new mathematical model for relative quantification in real-time RT-PCR. , 2001, Nucleic acids research.
[189] Jerilyn A. Walker,et al. Preparation of PCR-quality mouse genomic DNA with hot sodium hydroxide and tris (HotSHOT). , 2000, BioTechniques.
[190] J. Hodges,et al. Frontotemporal dementia , 1999, Neurology.
[191] S. Tonegawa,et al. Hippocampal lesions impair contextual fear conditioning in two strains of mice. , 1996, Behavioral neuroscience.
[192] Joseph E LeDoux,et al. Differential contribution of amygdala and hippocampus to cued and contextual fear conditioning. , 1992, Behavioral neuroscience.
[193] OUP accepted manuscript , 2022, Brain Communications.
[194] J. Deragon,et al. Moderated estimation of fold change and dispersion for RNA-seq data , 2020 .
[195] R Core Team,et al. R: A language and environment for statistical computing. , 2014 .
[196] G. Quirk,et al. Dissociable Roles of Prelimbic and Infralimbic Cortices, Ventral Hippocampus, and Basolateral Amygdala in the Expression and Extinction of Conditioned Fear , 2011, Neuropsychopharmacology.
[197] Buccafusco Jj. Cued and Contextual Fear Conditioning for Rodents -- Methods of Behavior Analysis in Neuroscience , 2009 .
[198] K. Browman,et al. Cued and Contextual Fear Conditioning for Rodents , 2009 .
[199] C. Walsh,et al. Mutations in ARFGEF2 implicate vesicle trafficking in neural progenitor proliferation and migration in the human cerebral cortex , 2004, Nature Genetics.
[200] H. Magoun,et al. INTO THE CENTRAL NERVOUS SYSTEM , 2003 .
[201] Harry T. Orr,et al. Identification and characterization of the gene causing type 1 spinocerebellar ataxia , 1994, Nature Genetics.