APOE3-R136S mutation confers resilience against tau pathology via cGAS-STING-IFN inhibition

The Christchurch mutation (R136S) on the APOE3 (E3S/S) gene is associated with low tau pathology and slowdown of cognitive decline despite the causal PSEN1 mutation and high levels of amyloid beta pathology in the carrier1. However, the molecular effects enabling E3S/S mutation to confer protection remain unclear. Here, we replaced mouse Apoe with wild-type human E3 or E3S/S on a tauopathy background. The R136S mutation markedly mitigated tau load and protected against tau-induced synaptic loss, myelin loss, and spatial learning. Additionally, the R136S mutation reduced microglial interferon response to tau pathology both in vivo and in vitro, suppressing cGAS-STING activation. Treating tauopathy mice carrying wild-type E3 with cGAS inhibitor protected against tau-induced synaptic loss and induced similar transcriptomic alterations to those induced by the R136S mutation across brain cell types. Thus, cGAS-STING-IFN inhibition recapitulates the protective effects of R136S against tauopathy. One-sentence summary The R136S mutation on APOE3 enhances resistance to tau-related disease processes by downregulating the cGAS-STING-IFN signaling pathway.

[1]  J. Fahel,et al.  Blockade of mGluR5 in astrocytes derived from human iPSCs modulates astrocytic function and increases phagocytosis , 2023, Frontiers in immunology.

[2]  J. Ulrich,et al.  APOE3ch alters microglial response and suppresses Aβ-induced tau seeding and spread , 2023, Cell.

[3]  Brian P. Grone,et al.  The APOE-R136S mutation protects against APOE4-driven Tau pathology, neurodegeneration and neuroinflammation , 2023, Nature Neuroscience.

[4]  S. DeKosky,et al.  Genome-wide analysis identifies novel loci influencing plasma apolipoprotein E concentration and Alzheimer’s disease risk , 2023, Molecular psychiatry.

[5]  Li Li,et al.  Astrocytic response mediated by the CLU risk allele inhibits OPC proliferation and myelination in a human iPSC model , 2023, Cell reports.

[6]  M. Takatama,et al.  Plasma ApoE4 Levels Are Lower than ApoE2 and ApoE3 Levels, and Not Associated with Plasma Aβ 40/42 Ratio as a Biomarker of Amyloid-β Amyloidosis in Alzheimer's Disease. , 2023, Journal of Alzheimer's disease : JAD.

[7]  D. Holtzman,et al.  Microglia-mediated T cell infiltration drives neurodegeneration in tauopathy , 2023, Nature.

[8]  B. Bacskai,et al.  Reduced excitatory neuron activity and interneuron-type-specific deficits in a mouse model of Alzheimer’s disease , 2022, Communications Biology.

[9]  Y. Asmann,et al.  Opposing effects of apoE2 and apoE4 on microglial activation and lipid metabolism in response to demyelination , 2022, Molecular Neurodegeneration.

[10]  William T. Ralvenius,et al.  APOE4 impairs myelination via cholesterol dysregulation in oligodendrocytes , 2022, Nature.

[11]  Justin S. Sanchez,et al.  Distinct tau neuropathology and cellular profiles of an APOE3 Christchurch homozygote protected against autosomal dominant Alzheimer’s dementia , 2022, Acta Neuropathologica.

[12]  D. Harvey,et al.  High-Density Lipoprotein Changes in Alzheimer’s Disease Are APOE Genotype-Specific , 2022, Biomedicines.

[13]  G. Rabinovici,et al.  Tau Beats Amyloid in Predicting Brain Atrophy in Alzheimer Disease: Implications for Prognosis and Clinical Trials , 2022, The Journal of Nuclear Medicine.

[14]  Holden C. Williams,et al.  APOE modulates microglial immunometabolism in response to age, amyloid pathology, and inflammatory challenge , 2022, bioRxiv.

[15]  Virginia M. Y. Lee,et al.  AD-linked R47H-TREM2 mutation induces disease-enhancing microglial states via AKT hyperactivation , 2021, Science Translational Medicine.

[16]  C. Jack,et al.  APOE3-Jacksonville (V236E) variant reduces self-aggregation and risk of dementia , 2021, Science Translational Medicine.

[17]  Gonçalo Lopes,et al.  New Open-Source Tools: Using Bonsai for Behavioral Tracking and Closed-Loop Experiments , 2021, Frontiers in Behavioral Neuroscience.

[18]  Hui Zheng,et al.  Clusterin secreted from astrocyte promotes excitatory synaptic transmission and ameliorates Alzheimer’s disease neuropathology , 2021, Molecular neurodegeneration.

[19]  V. Janout,et al.  Single Nucleotide Polymorphism rs11136000 of CLU Gene (Clusterin, ApoJ) and the Risk of Late-Onset Alzheimer’s Disease in a Central European Population , 2020, Neurochemical Research.

[20]  P. Giussani,et al.  The role of Sphingolipids in myelination and myelin stability and their involvement in childhood and adult demyelinating disorders , 2020, Journal of neurochemistry.

[21]  Yilan Xu,et al.  GABAergic Inhibitory Interneuron Deficits in Alzheimer’s Disease: Implications for Treatment , 2020, Frontiers in Neuroscience.

[22]  B. McEwen,et al.  Divergent roles of astrocytic versus neuronal EAAT2 deficiency on cognition and overlap with aging and Alzheimer’s molecular signatures , 2019, Proceedings of the National Academy of Sciences.

[23]  P. Scheltens,et al.  Early restoration of parvalbumin interneuron activity prevents memory loss and network hyperexcitability in a mouse model of Alzheimer’s disease , 2019, Molecular Psychiatry.

[24]  Seungpyo Hong,et al.  Diagnosis of Alzheimer’s disease utilizing amyloid and tau as fluid biomarkers , 2019, Experimental & Molecular Medicine.

[25]  Matthias Bethge,et al.  DeepLabCut: markerless pose estimation of user-defined body parts with deep learning , 2018, Nature Neuroscience.

[26]  James E. Goldman,et al.  White matter changes in Alzheimer’s disease: a focus on myelin and oligodendrocytes , 2018, Acta Neuropathologica Communications.

[27]  A. Fagan,et al.  ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy , 2017, Nature.

[28]  Markus Glatzel,et al.  The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. , 2017, Immunity.

[29]  I. Amit,et al.  A Unique Microglia Type Associated with Restricting Development of Alzheimer’s Disease , 2017, Cell.

[30]  Shay Ohayon,et al.  Open Ephys: an open-source, plugin-based platform for multichannel electrophysiology , 2017, Journal of neural engineering.

[31]  Geoffrey M. Barrett,et al.  Tau Pathology Induces Excitatory Neuron Loss, Grid Cell Dysfunction, and Spatial Memory Deficits Reminiscent of Early Alzheimer’s Disease , 2017, Neuron.

[32]  E. Boyden,et al.  Gamma frequency entrainment attenuates amyloid load and modifies microglia , 2016, Nature.

[33]  Jennifer Luebke,et al.  Depletion of microglia and inhibition of exosome synthesis halt tau propagation , 2015, Nature Neuroscience.

[34]  Arthur Konnerth,et al.  Neuronal hyperactivity – A key defect in Alzheimer's disease? , 2015, BioEssays : news and reviews in molecular, cellular and developmental biology.

[35]  C. Glass,et al.  Reducing macrophage proteoglycan sulfation increases atherosclerosis and obesity through enhanced type I interferon signaling. , 2014, Cell metabolism.

[36]  G. Bloom Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. , 2014, JAMA neurology.

[37]  K. Seyb,et al.  Small-molecule activator of glutamate transporter EAAT2 translation provides neuroprotection. , 2014, The Journal of clinical investigation.

[38]  C. DeCarli,et al.  Myelin injury and degraded myelin vesicles in Alzheimer's disease. , 2014, Current Alzheimer research.

[39]  Yanhui Liu,et al.  Identification of Tmem10 as a Novel Late-stage Oligodendrocytes Marker for Detecting Hypomyelination , 2013, International journal of biological sciences.

[40]  Edward O. Mann,et al.  Inhibitory Interneuron Deficit Links Altered Network Activity and Cognitive Dysfunction in Alzheimer Model , 2012, Cell.

[41]  Marcel Martin Cutadapt removes adapter sequences from high-throughput sequencing reads , 2011 .

[42]  L. Mucke,et al.  Amyloid-β–induced neuronal dysfunction in Alzheimer's disease: from synapses toward neural networks , 2010, Nature Neuroscience.

[43]  B. Hemmings,et al.  Carboxy-Terminal Modulator Protein (CTMP) is a mitochondrial protein that sensitizes cells to apoptosis. , 2009, Cellular signalling.

[44]  Noo Li Jeon,et al.  Presynaptic Regulation of Astroglial Excitatory Neurotransmitter Transporter GLT1 , 2009, Neuron.

[45]  M. Pangalos,et al.  Impact of Apolipoprotein E (ApoE) Polymorphism on Brain ApoE Levels , 2008, The Journal of Neuroscience.

[46]  Y. Uchijima,et al.  Carboxy-terminal modulator protein induces Akt phosphorylation and activation, thereby enhancing antiapoptotic, glycogen synthetic, and glucose uptake pathways. , 2007, American journal of physiology. Cell physiology.

[47]  R. Mahley,et al.  Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including Alzheimer's disease. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[48]  A. Fagan,et al.  Apolipoprotein E isoform-dependent amyloid deposition and neuritic degeneration in a mouse model of Alzheimer's disease. , 2000, Proceedings of the National Academy of Sciences of the United States of America.

[49]  Hugo Vanderstichele,et al.  Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein , 1998, Nature.

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

[51]  J. Hardy,et al.  The Amyloid Hypothesis of Alzheimer ’ s Disease : Progress and Problems on the Road to Therapeutics , 2009 .

[52]  E. Masliah,et al.  Deficient glutamate transport is associated with neurodegeneration in Alzheimer's disease. , 1996, Annals of neurology.