Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease
暂无分享,去创建一个
Evan Z. Macosko | E. Macosko | V. Gazestani | Naeem M. Nadaf | Tushar Kamath | Abdulraouf Abdulraouf | S. J. Burris | Jonah Langlieb | Karol Balderrama | Charles R Vanderburg | Karol S. Balderrama | Charles R. Vanderburg
[1] Houeto Jean-Luc. [Parkinson's disease]. , 2022, La Revue du praticien.
[2] Kathleen M. Jagodnik,et al. Gene Set Knowledge Discovery with Enrichr , 2021, Current protocols.
[3] J. Mulder,et al. Distinct amyloid-β and tau-associated microglia profiles in Alzheimer’s disease , 2021, Acta Neuropathologica.
[4] Daniel J. Gaffney,et al. Genome-wide meta-analysis, fine-mapping, and integrative prioritization implicate new Alzheimer’s disease risk genes , 2021, Nature Genetics.
[5] Patricia A. Castruita,et al. p53 is a central regulator driving neurodegeneration caused by C9orf72 poly(PR) , 2021, Cell.
[6] Evan Z. Macosko,et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2 , 2020, Nature Biotechnology.
[7] David Kulp,et al. Innovations present in the primate interneuron repertoire , 2020, Nature.
[8] P. May,et al. Single-cell sequencing of the human midbrain reveals glial activation and a neuronal state specific to Parkinson's disease , 2020, medRxiv.
[9] C. Webber,et al. A single-cell atlas of the human substantia nigra reveals cell-specific pathways associated with neurological disorders , 2020, Nature Communications.
[10] D. Goldstein,et al. Elevated COUP-TFII expression in dopaminergic neurons accelerates the progression of Parkinson’s disease through mitochondrial dysfunction , 2020, PLoS genetics.
[11] Rafael A. Irizarry,et al. Robust decomposition of cell type mixtures in spatial transcriptomics , 2020, Nature Biotechnology.
[12] Hunna J. Watson,et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease , 2020, Nature Genetics.
[13] N. Neff,et al. Molecular characterization of selectively vulnerable neurons in Alzheimer’s Disease , 2020, Nature Neuroscience.
[14] C. Webber,et al. Human‐Specific Transcriptome of Ventral and Dorsal Midbrain Dopamine Neurons , 2020, Annals of neurology.
[15] R. Awatramani,et al. Classification of Midbrain Dopamine Neurons Using Single-Cell Gene Expression Profiling Approaches , 2020, Trends in Neurosciences.
[16] Daniel Osorio,et al. Systematic determination of the mitochondrial proportion in human and mice tissues for single-cell RNA sequencing data quality control , 2020, bioRxiv.
[17] Christopher D. Brown,et al. The GTEx Consortium atlas of genetic regulatory effects across human tissues , 2019, Science.
[18] Allan R. Jones,et al. Conserved cell types with divergent features in human versus mouse cortex , 2019, Nature.
[19] Richard Reynolds,et al. Neuronal vulnerability and multilineage diversity in multiple sclerosis , 2019, Nature.
[20] Evan Z. Macosko,et al. Single-Cell Multi-omic Integration Compares and Contrasts Features of Brain Cell Identity , 2019, Cell.
[21] Maximilian Haeussler,et al. Single-cell genomics identifies cell type–specific molecular changes in autism , 2019, Science.
[22] Manolis Kellis,et al. Single-cell transcriptomic analysis of Alzheimer’s disease , 2019, Nature.
[23] Evan Z. Macosko,et al. Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution , 2019, Science.
[24] Dheeraj Malhotra,et al. Altered human oligodendrocyte heterogeneity in multiple sclerosis , 2019, Nature.
[25] Trygve E Bakken,et al. Single-nucleus and single-cell transcriptomes compared in matched cortical cell types , 2018, PloS one.
[26] D. Korzhevskii,et al. Immunohistochemical Characteristics of Neurons in the Substantia Nigra of the Human Brain , 2018, Neuroscience and Behavioral Physiology.
[27] Fan Zhang,et al. Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.
[28] Kamil Slowikowski,et al. Mixed-effects association of single cells identifies an expanded effector CD4+ T cell subset in rheumatoid arthritis , 2018, Science Translational Medicine.
[29] John Hardy,et al. Selective vulnerability in neurodegenerative diseases , 2018, Nature Neuroscience.
[30] Evan Z. Macosko,et al. Molecular Diversity and Specializations among the Cells of the Adult Mouse Brain , 2018, Cell.
[31] Paul Hoffman,et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.
[32] I. Amit,et al. A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.
[33] Gerome Breen,et al. Genetic identification of brain cell types underlying schizophrenia , 2017, Nature Genetics.
[34] Evan Z. Macosko,et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types , 2017, Nature Genetics.
[35] A. Pitsillides,et al. Pathogenic LRRK2 variants are gain-of-function mutations that enhance LRRK2-mediated repression of β-catenin signaling , 2017, Molecular Neurodegeneration.
[36] Lars E. Borm,et al. Molecular Diversity of Midbrain Development in Mouse, Human, and Stem Cells , 2016, Cell.
[37] Tomas Bergman,et al. A PBX1 transcriptional network controls dopaminergic neuron development and is impaired in Parkinson's disease , 2016, The EMBO journal.
[38] N. Greig,et al. Dopaminergic neuron‐specific deletion of p53 gene is neuroprotective in an experimental Parkinson's disease model , 2016, Journal of neurochemistry.
[39] Grace X. Y. Zheng,et al. Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.
[40] P. Linsley,et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data , 2015, Genome Biology.
[41] C. Creighton,et al. Increased COUP-TFII expression in adult hearts induces mitochondrial dysfunction resulting in heart failure , 2015, Nature Communications.
[42] E. Arenas,et al. How to make a midbrain dopaminergic neuron , 2015, Development.
[43] Allon M. Klein,et al. Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells , 2015, Cell.
[44] Evan Z. Macosko,et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets , 2015, Cell.
[45] Joris M. Mooij,et al. MAGMA: Generalized Gene-Set Analysis of GWAS Data , 2015, PLoS Comput. Biol..
[46] Georg Auburger,et al. The Brainstem Pathologies of Parkinson's Disease and Dementia with Lewy Bodies , 2015, Brain pathology.
[47] P. Greengard,et al. Molecular determinants of selective dopaminergic vulnerability in Parkinson’s disease: an update , 2014, Front. Neuroanat..
[48] J. Kehr,et al. Sox6 and Otx2 control the specification of substantia nigra and ventral tegmental area dopamine neurons. , 2014, Cell reports.
[49] A. Björklund,et al. NURR1 in Parkinson disease—from pathogenesis to therapeutic potential , 2013, Nature Reviews Neurology.
[50] O. Isacson,et al. Functional enhancement and protection of dopaminergic terminals by RAB3B overexpression , 2009, Proceedings of the National Academy of Sciences.
[51] Patrick M. Abou-Sleiman,et al. Transcriptional repression of p53 by parkin and impairment by mutations associated with autosomal recessive juvenile Parkinson's disease , 2009, Nature Cell Biology.
[52] E. Hirsch,et al. Regional vulnerability of mesencephalic dopaminergic neurons prone to degenerate in Parkinson's disease: A post-mortem study in human control subjects , 2006, Neurobiology of Disease.
[53] Pablo Tamayo,et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.
[54] S. Snyder,et al. p53 Mediates Cellular Dysfunction and Behavioral Abnormalities in Huntington’s Disease , 2005, Neuron.
[55] A. Graybiel,et al. The substantia nigra of the human brain. I. Nigrosomes and the nigral matrix, a compartmental organization based on calbindin D(28K) immunohistochemistry. , 1999, Brain : a journal of neurology.
[56] A. Graybiel,et al. The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson's disease. , 1999, Brain : a journal of neurology.
[57] Y. Kitamura,et al. Changes of p53 in the brains of patients with Alzheimer's disease. , 1997, Biochemical and biophysical research communications.
[58] D. Riche,et al. Chronic MPTP treatment reproduces in baboons the differential vulnerability of mesencephalic dopaminergic neurons observed in parkinson's disease , 1994, Neuroscience.
[59] E. Rosengren,et al. Isolation of a new tautomerase monitored by the conversion of D-dopachrome to 5,6-dihydroxyindole. , 1993, Biochemical and biophysical research communications.
[60] A. Lees,et al. Ageing and Parkinson's disease: substantia nigra regional selectivity. , 1991, Brain : a journal of neurology.
[61] W. Gibb,et al. Anatomy, pigmentation, ventral and dorsal subpopulations of the substantia nigra, and differential cell death in Parkinson's disease. , 1991, Journal of neurology, neurosurgery, and psychiatry.
[62] P. Mcgeer,et al. Relative sparing in Parkinson's disease of substantia nigra dopamine neurons containing calbindin-D28K , 1990, Brain Research.
[63] C. Markham,et al. Selective loss of subpopulations of ventral mesencephalic dopaminergic neurons in the monkey following exposure to MPTP , 1987, Brain Research.