PLCG2 protective variant p.P522R modulates tau pathology and disease progression in patients with mild cognitive impairment.


Journal

Acta neuropathologica
ISSN: 1432-0533
Titre abrégé: Acta Neuropathol
Pays: Germany
ID NLM: 0412041

Informations de publication

Date de publication:
06 2020
Historique:
received: 09 11 2019
accepted: 20 02 2020
revised: 19 02 2020
pubmed: 14 3 2020
medline: 9 6 2021
entrez: 14 3 2020
Statut: ppublish

Résumé

A rare coding variant (rs72824905, p.P522R) conferring protection against Alzheimer's disease (AD) was identified in the gene encoding the enzyme phospholipase-C-γ2 (PLCG2) that is highly expressed in microglia. To explore the protective nature of this variant, we employed latent process linear mixed models to examine the association of p.P522R with longitudinal cognitive decline in 3595 MCI patients, and in 10,097 individuals from population-based studies. Furthermore, association with CSF levels of pTau

Identifiants

pubmed: 32166339
doi: 10.1007/s00401-020-02138-6
pii: 10.1007/s00401-020-02138-6
pmc: PMC7244617
doi:

Substances chimiques

Amyloid beta-Peptides 0
Biomarkers 0
tau Proteins 0
PLCG2 protein, human EC 3.1.4.3
Phospholipase C gamma EC 3.1.4.3

Types de publication

Journal Article Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov't Research Support, U.S. Gov't, Non-P.H.S.

Langues

eng

Sous-ensembles de citation

IM

Pagination

1025-1044

Subventions

Organisme : NIA NIH HHS
ID : U01 AG024904
Pays : United States
Organisme : HSRD VA
ID : CDA 10-014
Pays : United States

Investigateurs

Luca Kleineidam (L)
Ilker Karaca (I)
Michael T Heneka (MT)
Wolfgang Maier (W)
Anja Schneider (A)
Michael Wagner (M)
Vincent Chouraki (V)
Phillipe Amouyel (P)
Jean-Charles Lambert (JC)
Tomasz Próchnicki (T)
Eicke Latz (E)
Sven J van der Lee (SJ)
Iris E Jansen (IE)
Marc Hulsman (M)
Philip Scheltens (P)
Wiesje M van der Flier (WM)
Henne Holstege (H)
Laura Madrid-Márquez (L)
Antonio González-Pérez (A)
Mª Eugenia Sáez (ME)
Holger Wagner-Thelen (H)
Pamela V Martino Adami (PV)
Frank Jessen (F)
Alfredo Ramirez (A)
Leonie Weinhold (L)
Matthias Schmid (M)
Steffen Wolfsgruber (S)
Frederic Brosseron (F)
Anne Boland (A)
Jean-Francois Deleuze (JF)
Piotr Lewczuk (P)
Johannes Kornhuber (J)
Julius Popp (J)
Oliver Peters (O)
Claudine Berr (C)
Reinhard Heun (R)
Lutz Frölich (L)
Christophe Tzourio (C)
Jean-François Dartigues (JF)
Michael Hüll (M)
Ana Espinosa (A)
Isabel Hernández (I)
Itziar de Rojas (I)
Adelina Orellana (A)
Sergi Valero (S)
Agustin Ruiz (A)
Lluis Tarraga (L)
Merce Boada (M)
Najada Stringa (N)
Natasja M van Schoor (NM)
Martijn Huisman (M)
Eckart Rüther (E)
Jens Wiltfang (J)
Martin Scherer (M)
Steffi Riedel-Heller (S)

Références

Abdelnour C, van Steenoven I, Londos E et al (2016) Alzheimer’s disease cerebrospinal fluid biomarkers predict cognitive decline in lewy body dementia. Mov Disord 31(8):1203–1208
pubmed: 27296778 doi: 10.1002/mds.26668
Abner EL, Kryscio RJ, Schmitt FA et al (2017) Outcomes after diagnosis of mild cognitive impairment in a large autopsy series. Ann Neurol 81(4):549–559
pubmed: 28224671 pmcid: 5401633 doi: 10.1002/ana.24903
Arboleda-Velasquez JF, Lopera F, O’Hare M et al (2019) Resistance to autosomal dominant Alzheimer’s disease in an APOE3 Christchurch homozygote: a case report. Nat Med 25(11):1680–1683
pubmed: 31686034 pmcid: 6898984 doi: 10.1038/s41591-019-0611-3
Asai H, Ikezu S, Tsunoda S et al (2015) Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 18(11):1584–1593
pubmed: 26436904 pmcid: 4694577 doi: 10.1038/nn.4132
Bhaskar K, Konerth M, Kokiko-Cochran ON et al (2010) Regulation of tau pathology by the microglial fractalkine receptor. Neuron 68(1):19–31
pubmed: 20920788 pmcid: 2950825 doi: 10.1016/j.neuron.2010.08.023
Boyle PA, Wilson RS, Yu L et al (2013) Much of late life cognitive decline is not due to common neurodegenerative pathologies. Ann Neurol 74(3):478–489
pubmed: 23798485 doi: 10.1002/ana.23964
Braak H, Thal DR, Ghebremedhin E et al (2011) Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol 70(11):960–969
doi: 10.1097/NEN.0b013e318232a379 pubmed: 22002422
Chae JJ, Park YH, Park C et al (2015) Brief report: connecting two pathways through Ca2
pubmed: 25418813 pmcid: 4369162 doi: 10.1002/art.38961
Cohen G, Makranz C, Spira M et al (2006) Non-PKC DAG/Phorbol-Ester receptor (s) inhibit complement receptor-3 and nPKC inhibit scavenger receptor-AI/II-mediated myelin phagocytosis but cPKC, PI3k, and PLCγ activate myelin phagocytosis by both. Glia 53(5):538–550
pubmed: 16374778 doi: 10.1002/glia.20304
Conway OJ, Carrasquillo MM, Wang X et al (2018) ABI3 and PLCG2 missense variants as risk factors for neurodegenerative diseases in Caucasians and African Americans. Mol Neurodegener 13(1):53
pubmed: 30326945 pmcid: 6190665 doi: 10.1186/s13024-018-0289-x
Curran PJ, Hussong AM (2009) Integrative data analysis: the simultaneous analysis of multiple data sets. Psychol Methods 14(2):81
pubmed: 19485623 pmcid: 19485623 doi: 10.1037/a0015914
Dagher NN, Najafi AR, Kayala KMN et al (2015) Colony-stimulating factor 1 receptor inhibition prevents microglial plaque association and improves cognition in 3xTg-AD mice. J Neuroinflamm 12(1):139
doi: 10.1186/s12974-015-0366-9
Dalmasso MC, Brusco LI, Olivar N et al (2019) Transethnic meta-analysis of rare coding variants in PLCG2, ABI3, and TREM2 supports their general contribution to Alzheimer’s disease. Transl Psychiatry 9(1):55
pubmed: 30705288 pmcid: 6355764 doi: 10.1038/s41398-019-0394-9
Deczkowska A, Keren-Shaul H, Weiner A et al (2018) Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell 173(5):1073–1081
pubmed: 29775591 doi: 10.1016/j.cell.2018.05.003
Enders CK (2010) Applied missing data analysis. Guilford Press, New York
Ferrari R, Wang Y, Vandrovcova J et al (2017) Genetic architecture of sporadic frontotemporal dementia and overlap with Alzheimer’s and Parkinson’s diseases. J Neurol Neurosurg Psychiatry 88(2):152–164
pubmed: 27899424 doi: 10.1136/jnnp-2016-314411
Finan C, Gaulton A, Kruger FA et al (2017) The druggable genome and support for target identification and validation in drug development. Sci Transl Med 9(383):eaag1166
pubmed: 28356508 pmcid: 6321762 doi: 10.1126/scitranslmed.aag1166
Fjell AM, McEvoy L, Holland D et al (2014) What is normal in normal aging? Effects of aging, amyloid and Alzheimer’s disease on the cerebral cortex and the hippocampus. Prog Neurobiol 117:20–40
pubmed: 24548606 pmcid: 4343307 doi: 10.1016/j.pneurobio.2014.02.004
Friedman BA, Srinivasan K, Ayalon G et al (2018) Diverse brain myeloid expression profiles reveal distinct microglial activation states and aspects of Alzheimer’s disease not evident in mouse models. Cell Rep 22(3):832–847
pubmed: 29346778 doi: 10.1016/j.celrep.2017.12.066
Guerreiro RJ, Lohmann E, Brás JM et al (2013) Using exome sequencing to reveal mutations in TREM2 presenting as a frontotemporal dementia–like syndrome without bone involvement. JAMA Neurol 70(1):78–84
pubmed: 23318515 pmcid: 4001789 doi: 10.1001/jamaneurol.2013.579
Hammond TR, Marsh SE, Stevens B (2019) Immune signaling in neurodegeneration. Immunity 50(4):955–974
pubmed: 30995509 pmcid: 6822103 doi: 10.1016/j.immuni.2019.03.016
Haukedal H, Freude K (2019) Implications of microglia in amyotrophic lateral sclerosis and frontotemporal dementia. J Mol Biol 431(9):1818–1829
pubmed: 30763568 doi: 10.1016/j.jmb.2019.02.004
Hayat SA, Luben R, Dalzell N et al (2018) Understanding the relationship between cognition and death: a within cohort examination of cognitive measures and mortality. Eur J Epidemiol 33(11):1049–1062
pubmed: 30203336 pmcid: 6208995 doi: 10.1007/s10654-018-0439-z
Heckman MG, Kasanuki K, Brennan RR et al (2019) Association of MAPT H1 subhaplotypes with neuropathology of lewy body disease. Mov Disord 34:1325–1332
pubmed: 31234228 doi: 10.1002/mds.27773
Herrup K (2010) Reimagining Alzheimer’s disease—an age-based hypothesis. J Neurosci 30(50):16755–16762
pubmed: 21159946 pmcid: 3004746 doi: 10.1523/JNEUROSCI.4521-10.2010
Hohman TJ, Tommet D, Marks S et al (2017) Evaluating Alzheimer’s disease biomarkers as mediators of age-related cognitive decline. Neurobiol Aging 58:120–128
pubmed: 28732249 pmcid: 5710827 doi: 10.1016/j.neurobiolaging.2017.06.022
Hong S, Beja-Glasser VF, Nfonoyim BM et al (2016) Complement and microglia mediate early synapse loss in Alzheimer mouse models. Science 352(6286):712–716
pubmed: 27033548 pmcid: 5094372 doi: 10.1126/science.aad8373
Ising C, Venegas C, Zhang S et al (2019) NLRP3 inflammasome activation drives tau pathology. Nature 575(7784):669–673
pubmed: 31748742 doi: 10.1038/s41586-019-1769-z
Jack R Jr, Clifford, Bennett DA, Blennow K et al (2018) NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement 14(4):535–562
pubmed: 29653606 pmcid: 5958625 doi: 10.1016/j.jalz.2018.02.018
Jack CR Jr, Clifford R, Knopman DS, Jagust WJ et al (2013) Tracking pathophysiological processes in Alzheimer’s disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol 12(2):207–216
pubmed: 23332364 pmcid: 3622225 doi: 10.1016/S1474-4422(12)70291-0
Jagust W (2013) Vulnerable neural systems and the borderland of brain aging and neurodegeneration. Neuron 77(2):219–234
pubmed: 23352159 pmcid: 3558930 doi: 10.1016/j.neuron.2013.01.002
Jansen WJ, Ossenkoppele R, Knol DL et al (2015) Prevalence of cerebral amyloid pathology in persons without dementia: a meta-analysis. Jama 313(19):1924–1938
pubmed: 25988462 pmcid: 4486209 doi: 10.1001/jama.2015.4668
Jansen IE, Savage JE, Watanabe K et al (2019) Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat Genet 51:404–413
pubmed: 30617256 pmcid: 6836675 doi: 10.1038/s41588-018-0311-9
Jay TR, von Saucken VE, Landreth GE (2017) TREM2 in neurodegenerative diseases. Mol Neurodegener 12(1):56
pubmed: 28768545 pmcid: 5541421 doi: 10.1186/s13024-017-0197-5
Jones L, Lambert J-C, Wang L-S et al (2015) Convergent genetic and expression data implicate immunity in Alzheimer’s disease. Alzheimers Dement 11(6):658–671
doi: 10.1016/j.jalz.2014.05.1757
Jun G, Ibrahim-Verbaas CA, Vronskaya M et al (2016) A novel Alzheimer disease locus located near the gene encoding tau protein. Mol Psychiatry 21(1):108
pubmed: 25778476 doi: 10.1038/mp.2015.23
Kamb A, Harper S, Stefansson K (2013) Human genetics as a foundation for innovative drug development. Nat Biotechnol 31(11):975
pubmed: 24213769 pmcid: 24213769 doi: 10.1038/nbt.2732
Karran E, Mercken M, de Strooper B (2011) The amyloid cascade hypothesis for Alzheimer’s disease: an appraisal for the development of therapeutics. Nat Rev Drug Discov 10(9):698
pubmed: 21852788 doi: 10.1038/nrd3505
Keren-Shaul H, Spinrad A, Weiner A et al (2017) A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169(7):1276–1290
pubmed: 28602351 doi: 10.1016/j.cell.2017.05.018 pmcid: 28602351
Koller M, Stahel WA (2017) Nonsingular subsampling for regression S estimators with categorical predictors. Comput Stat 32(2):631–646
doi: 10.1007/s00180-016-0679-x
Kunkle BW, Grenier-Boley B, Sims R et al (2019) Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat Genet 51(3):414–430
pubmed: 30820047 pmcid: 6463297 doi: 10.1038/s41588-019-0358-2
Lacour A, Espinosa A, Louwersheimer E et al (2017) Genome-wide significant risk factors for Alzheimer’s disease: role in progression to dementia due to Alzheimer’s disease among subjects with mild cognitive impairment. Mol Psychiatry 22(1):153
pubmed: 26976043 doi: 10.1038/mp.2016.18
Lall D, Baloh RH (2017) Microglia and C9orf72 in neuroinflammation and ALS and frontotemporal dementia. J Clin Invest 127(9):3250–3258
pubmed: 28737506 pmcid: 5669558 doi: 10.1172/JCI90607
Lee CD, Daggett A, Gu X et al (2018) Elevated TREM2 gene dosage reprograms microglia responsivity and ameliorates pathological phenotypes in Alzheimer’s disease models. Neuron 97(5):1032–1048
pubmed: 29518357 pmcid: 5927822 doi: 10.1016/j.neuron.2018.02.002
Liao Y, Wang J, Jaehnig EJ et al (2019) WebGestalt 2019: gene set analysis toolkit with revamped UIs and APIs. Nucleic Acids Res 47(W1):W100–W205
doi: 10.1093/nar/gkz401
Lill CM, Rengmark A, Pihlstrøm L et al (2015) The role of TREM2 R47H as a risk factor for Alzheimer’s disease, frontotemporal lobar degeneration, amyotrophic lateral sclerosis, and Parkinson’s disease. Alzheimers Dement 11(12):1407–1416
pubmed: 25936935 pmcid: 4627856 doi: 10.1016/j.jalz.2014.12.009
Magno L, Lessard CB, Martins M et al (2019) Alzheimer’s disease phospholipase C-gamma-2 (PLCG2) protective variant is a functional hypermorph. Alzheimers Res Ther 11(1):16
pubmed: 30711010 pmcid: 6359863 doi: 10.1186/s13195-019-0469-0
Makranz C, Cohen G, Baron A et al (2004) Phosphatidylinositol 3-kinase, phosphoinositide-specific phospholipase-Cγ and protein kinase-C signal myelin phagocytosis mediated by complement receptor-3 alone and combined with scavenger receptor-AI/II in macrophages. Neurobiol Dis 15(2):279–286
pubmed: 15006698 doi: 10.1016/j.nbd.2003.11.007
Mathys H, Davila-Velderrain J, Peng Z et al (2019) Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570(7761):332–337
pubmed: 31042697 pmcid: 6865822 doi: 10.1038/s41586-019-1195-2
Muthén LK, Muthén BO (1998–2012). Mplus user’s guide, 7th Edn. Muthén & Muthén, Los Angeles, CA
Olmos-Alonso A, Schetters STT, Sri S et al (2016) Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain 139(3):891–907
pubmed: 26747862 pmcid: 4766375 doi: 10.1093/brain/awv379
Ombrello MJ, Remmers EF, Sun G et al (2012) Cold urticaria, immunodeficiency, and autoimmunity related to PLCG2 deletions. N Engl J Med 366(4):330–338
pubmed: 22236196 pmcid: 3298368 doi: 10.1056/NEJMoa1102140
Ossenkoppele R, Jansen WJ, Rabinovici GD et al (2015) Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. Jama 313(19):1939–1950
pubmed: 25988463 pmcid: 4517678 doi: 10.1001/jama.2015.4669
Peng Q, Malhotra S, Torchia JA et al (2010) TREM2-and DAP12-dependent activation of PI3K requires DAP10 and is inhibited by SHIP1. Sci Signal 3(122):ra38–ra38
pubmed: 20484116 pmcid: 2900152 doi: 10.1126/scisignal.2000500
Plenge RM, Scolnick EM, Altshuler D (2013) Validating therapeutic targets through human genetics. Nat Rev Drug Discov 12(8):581
pubmed: 23868113 doi: 10.1038/nrd4051
Proust-Lima C, Dartigues J-F, Jacqmin-Gadda H (2011) Misuse of the linear mixed model when evaluating risk factors of cognitive decline. Am J Epidemiol 174(9):1077–1088
pubmed: 21965187 pmcid: 3551607 doi: 10.1093/aje/kwr243
Proust-Lima C, Philipps V, Liquet B (2017) Estimation of extended mixed models using latent classes and latent processes: The R package lcmm. J Stat Softw 78(2):56. https://doi.org/10.18637/jss.v078.i02
doi: 10.18637/jss.v078.i02
R Core Team (2018) R: a language and environment for statistical computing. R Foundation for Statistical Computing, Vienna. http://www.R-project.org
Raghavan N, Samtani MN, Farnum M et al (2013) The ADAS-Cog revisited: novel composite scales based on ADAS-Cog to improve efficiency in MCI and early AD trials. Alzheimers Dement 9(1):S21–S31
pubmed: 23127469 doi: 10.1016/j.jalz.2012.05.2187
Rauchmann B-S, Schneider-Axmann T, Alexopoulos P et al (2019) CSF soluble TREM2 as a measure of immune response along the Alzheimer’s disease continuum. Neurobiol Aging 74:182–190
pubmed: 30458365 doi: 10.1016/j.neurobiolaging.2018.10.022
Rayaprolu S, Mullen B, Baker M et al (2013) TREM2 in neurodegeneration: evidence for association of the p R47H variant with frontotemporal dementia and Parkinson’s disease. Mol Neurodegener 8(1):19
pubmed: 23800361 pmcid: 3691612 doi: 10.1186/1750-1326-8-19
Roberts R, Knopman DS (2013) Classification and epidemiology of MCI. Clin Geriatr Med 29(4):753–772
pubmed: 24094295 doi: 10.1016/j.cger.2013.07.003
Roberts RO, Aakre JA, Kremers WK et al (2018) Prevalence and outcomes of amyloid positivity among persons without dementia in a longitudinal, population-based setting. JAMA Neurol 75(8):970–979
pubmed: 29710225 pmcid: 6142936 doi: 10.1001/jamaneurol.2018.0629
Rongve A, Witoelar A, Ruiz A et al (2019) GBA and APOE ε4 associate with sporadic dementia with Lewy bodies in European genome wide association study. Sci Rep 9(1):7013
pubmed: 31065058 pmcid: 6504850 doi: 10.1038/s41598-019-43458-2
Schulze-Luehrmann J, Ghosh S (2006) Antigen-receptor signaling to nuclear factor κB. Immunity 25(5):701–715
pubmed: 17098202 doi: 10.1016/j.immuni.2006.10.010
Shen M-Y, Hsiao G, Fong T-H et al (2008) Expression of amyloid beta peptide in human platelets: pivotal role of the phospholipase Cγ2-protein kinase C pathway in platelet activation. Pharmacol Res 57(2):151–158
pubmed: 18313326 doi: 10.1016/j.phrs.2008.01.004
Shi Y, Holtzman DM (2018) Interplay between innate immunity and Alzheimer disease: APOE and TREM2 in the spotlight. Nat Rev Immunol 18(12):759–772
pubmed: 30140051 pmcid: 6425488 doi: 10.1038/s41577-018-0051-1
Sims R, van der Lee SJ, Naj AC et al (2017) Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet 49(9):1373
pubmed: 28714976 pmcid: 5669039 doi: 10.1038/ng.3916
Song WM, Joshita S, Zhou Y et al (2018) Humanized TREM2 mice reveal microglia-intrinsic and-extrinsic effects of R47H polymorphism. J Exp Med 215(3):745–760
pubmed: 29321225 pmcid: 5839761 doi: 10.1084/jem.20171529
Sosna J, Philipp S, Albay R et al (2018) Early long-term administration of the CSF1R inhibitor PLX3397 ablates microglia and reduces accumulation of intraneuronal amyloid, neuritic plaque deposition and pre-fibrillar oligomers in 5XFAD mouse model of Alzheimer’s disease. Mol Neurodegener 13(1):11
pubmed: 29490706 pmcid: 5831225 doi: 10.1186/s13024-018-0244-x
Spangenberg EE, Lee RJ, Najafi AR et al (2016) Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-β pathology. Brain 139(4):1265–1281
pubmed: 26921617 pmcid: 5006229 doi: 10.1093/brain/aww016
Spangenberg E, Severson PL, Hohsfield LA et al (2019) Sustained microglial depletion with CSF1R inhibitor impairs parenchymal plaque development in an Alzheimer’s disease model. Nat Commun 10(1):1–21
doi: 10.1038/s41467-019-11674-z
Stancu I-C, Cremers N, Vanrusselt H et al (2019) Aggregated Tau activates NLRP3–ASC inflammasome exacerbating exogenously seeded and non-exogenously seeded Tau pathology in vivo. Acta Neuropathol 137(4):599–617
pubmed: 30721409 pmcid: 6426830 doi: 10.1007/s00401-018-01957-y
Suárez-Calvet M, Morenas-Rodríguez E, Kleinberger G et al (2019) Early increase of CSF sTREM2 in Alzheimer’s disease is associated with tau related-neurodegeneration but not with amyloid-β pathology. Mol Neurodegener 14(1):1
pubmed: 30630532 pmcid: 6327425 doi: 10.1186/s13024-018-0301-5
Szklarczyk D, Gable AL, Lyon D et al (2018) STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res 47(D1):D607–D613
pmcid: 6323986 doi: 10.1093/nar/gky1131
Terry RD, Masliah E, Salmon DP et al (1991) Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol 30(4):572–580
pubmed: 1789684 doi: 10.1002/ana.410300410
van Dam S, Cordeiro R, Craig T et al (2012) GeneFriends: an online co-expression analysis tool to identify novel gene targets for aging and complex diseases. BMC Genom 13(1):535
doi: 10.1186/1471-2164-13-535
van der Lee SJ, Conway OJ, Jansen I et al (2019) A nonsynonymous mutation in PLCG2 reduces the risk of Alzheimer’s disease, dementia with Lewy bodies and frontotemporal dementia, and increases the likelihood of longevity. Acta neuropathol 138:1–14
doi: 10.1007/s00401-019-02030-y
Venegas C, Kumar S, Franklin BS et al (2017) Microglia-derived ASC specks cross-seed amyloid-β in Alzheimer’s disease. Nature 552(7685):355–361
pubmed: 29293211 doi: 10.1038/nature25158
Waegaert R, Dirrig-Grosch S, Parisot F et al (2020) Longitudinal transcriptomic analysis of altered pathways in a CHMP2Bintron5-based model of ALS-FTD. Neurobiol Dis 136:104710
pubmed: 31837425 doi: 10.1016/j.nbd.2019.104710
Wood SN (2003) Thin plate regression splines. J R Stat Soc Series B Stat Methodol 65(1):95–114
doi: 10.1111/1467-9868.00374
Wood SN (2011) Fast stable restricted maximum likelihood and marginal likelihood estimation of semiparametric generalized linear models. J R Stat Soc Series B Stat Methodol 73(1):3–36
doi: 10.1111/j.1467-9868.2010.00749.x
Woollacott IOC, Nicholas JM, Heslegrave A et al (2018) Cerebrospinal fluid soluble TREM2 levels in frontotemporal dementia differ by genetic and pathological subgroup. Alzheimers Res Ther 10(1):79
pubmed: 30111356 pmcid: 6094471 doi: 10.1186/s13195-018-0405-8
Yeh FL, Wang Y, Tom I et al (2016) TREM2 binds to apolipoproteins, including APOE and CLU/APOJ, and thereby facilitates uptake of amyloid-beta by microglia. Neuron 91(2):328–340
pubmed: 27477018 doi: 10.1016/j.neuron.2016.06.015
Yoshiyama Y, Higuchi M, Zhang B et al (2007) Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53(3):337–351
pubmed: 17270732 doi: 10.1016/j.neuron.2007.01.010
Zanoni I, Ostuni R, Marek LR et al (2011) CD14 controls the LPS-induced endocytosis of Toll-like receptor 4. Cell 147(4):868–880
pubmed: 22078883 pmcid: 3217211 doi: 10.1016/j.cell.2011.09.051
Zanoni I, Tan Y, Di Gioia M et al (2017) By capturing inflammatory lipids released from dying cells, the receptor CD14 induces inflammasome-dependent phagocyte hyperactivation. Immunity 47(4):697–709
pubmed: 29045901 pmcid: 5747599 doi: 10.1016/j.immuni.2017.09.010
Zhang B, Gaiteri C, Bodea L-G et al (2013) Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer’s disease. Cell 153(3):707–720
pubmed: 23622250 pmcid: 3677161 doi: 10.1016/j.cell.2013.03.030
Zhou HH, Singh V, Johnson SC et al (2018) Statistical tests and identifiability conditions for pooling and analyzing multisite datasets. Proc Natl Acad Sci 115(7):1481–1486
pubmed: 29386387 doi: 10.1073/pnas.1719747115

Auteurs

Luca Kleineidam (L)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Vincent Chouraki (V)

Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France.
Epidemiology and Public Health Department, Centre Hospitalier Universitaire de Lille, Lille, France.

Tomasz Próchnicki (T)

Institute of Innate Immunity, University Hospitals Bonn, Bonn, Germany.

Sven J van der Lee (SJ)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Laura Madrid-Márquez (L)

Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain.

Holger Wagner-Thelen (H)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.

Ilker Karaca (I)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.

Leonie Weinhold (L)

Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany.

Steffen Wolfsgruber (S)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Anne Boland (A)

Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Évry, France.

Pamela V Martino Adami (PV)

Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.

Piotr Lewczuk (P)

Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.
Department of Neurodegeneration Diagnostics, Medical University of Białystok, Białystok, Poland.
Department of Biochemical Diagnostics, University Hospital of Białystok, Białystok, Poland.

Julius Popp (J)

Department of Psychiatry, Lausanne University Hospital, Prilly, Switzerland.
Department of Geriatric Psychiatry, University Hospital of Psychiatry Zurich, Zurich, Switzerland.

Frederic Brosseron (F)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Iris E Jansen (IE)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Marc Hulsman (M)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Johannes Kornhuber (J)

Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen, and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.

Oliver Peters (O)

Department of Psychiatry, Charité - Universitätsmedizin Berlin, Berlin, Germany.
DZNE, German Center for Neurodegenerative Diseases, Berlin, Germany.

Claudine Berr (C)

INSERM, University Montpellier, Neuropsychiatry: Epidemiological and Clinical Research, Montpellier, France.

Reinhard Heun (R)

Department of Psychiatry and Psychotherapy, University Hospital Bonn, 53127, Bonn, Germany.

Lutz Frölich (L)

Department of Geriatric Psychiatry, Medical Faculty Mannheim, Central Institute of Mental Health, University of Heidelberg, Mannheim, Germany.

Christophe Tzourio (C)

Inserm, Bordeaux Population Health Research Center, UMR1219, University of Bordeaux, Bordeaux, France.

Jean-François Dartigues (JF)

Inserm, Bordeaux Population Health Research Center, UMR1219, University of Bordeaux, Bordeaux, France.

Michael Hüll (M)

Department of Psychiatry and Psychotherapy, Center for Psychiatry, Clinic for Geriatric Psychiatry and Psychotherapy Emmendingen, University of Freiburg, Freiburg, Germany.

Ana Espinosa (A)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Isabel Hernández (I)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Itziar de Rojas (I)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Adelina Orellana (A)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.

Sergi Valero (S)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Najada Stringa (N)

Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Natasja M van Schoor (NM)

Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Martijn Huisman (M)

Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam UMC-Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Philip Scheltens (P)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Eckart Rüther (E)

Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.

Jean-Francois Deleuze (JF)

Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, Évry, France.

Jens Wiltfang (J)

Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany.
German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany.
iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal.

Lluis Tarraga (L)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Matthias Schmid (M)

German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
Institute of Medical Biometry, Informatics and Epidemiology, University Hospital of Bonn, Bonn, Germany.

Martin Scherer (M)

Department of Primary Medical Care, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.

Steffi Riedel-Heller (S)

Institute of Social Medicine, Occupational Health and Public Health, University of Leipzig, Leipzig, Germany.

Michael T Heneka (MT)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.

Philippe Amouyel (P)

Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France.

Frank Jessen (F)

German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany.

Merce Boada (M)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Wolfgang Maier (W)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Anja Schneider (A)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Antonio González-Pérez (A)

Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain.

Wiesje M van der Flier (WM)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Michael Wagner (M)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.

Jean-Charles Lambert (JC)

Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque Et déterminants moléculaires des maladies liées au vieillissement, Lille, France.

Henne Holstege (H)

Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
Department of Clinical Genetics, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.

Mª Eugenia Sáez (ME)

Andalusian Bioinformatics Research Centre (CAEBi), Seville, Spain.

Eicke Latz (E)

German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
Institute of Innate Immunity, University Hospitals Bonn, Bonn, Germany.
Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
Centre for Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, Trondheim, Norway.

Agustin Ruiz (A)

Research Center and Memory Clinic, Fundació ACE, Institut Català de Neurociències Aplicades-Universitat Internacional de Catalunya-Barcelona, Barcelona, Spain.
Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.

Alfredo Ramirez (A)

Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany. alfredo.ramirez@uk-koeln.de.
Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany. alfredo.ramirez@uk-koeln.de.

Articles similaires

[Redispensing of expensive oral anticancer medicines: a practical application].

Lisanne N van Merendonk, Kübra Akgöl, Bastiaan Nuijen
1.00
Humans Antineoplastic Agents Administration, Oral Drug Costs Counterfeit Drugs

Smoking Cessation and Incident Cardiovascular Disease.

Jun Hwan Cho, Seung Yong Shin, Hoseob Kim et al.
1.00
Humans Male Smoking Cessation Cardiovascular Diseases Female
Humans United States Aged Cross-Sectional Studies Medicare Part C
1.00
Humans Yoga Low Back Pain Female Male

Classifications MeSH