Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37577582

ABSTRACT

Background: Genetic study of late-onset Alzheimer's disease (AD) reveals that a rare Arginine-to-Histamine mutation at amino acid residue 47 (R47H) in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) results in increased disease risk. TREM2 plays critical roles in regulating microglial response to amyloid plaques in AD, leading to their clustering and activation surrounding the plaques. We previously showed that increasing human TREM2 gene dosage exerts neuroprotective effects against AD-related deficits in amyloid depositing mouse models of AD. However, the in vivo effects of the R47H mutation on human TREM2-mediated microglial reprogramming and neuroprotection remains poorly understood. Method: Here we created a BAC transgenic mouse model expressing human TREM2 with the R47H mutation in its cognate genomic context (BAC-TREM2-R47H). Importantly, the BAC used in this study was engineered to delete critical exons of other TREM-like genes on the BAC to prevent confounding effects of overexpressing multiple TREM-like genes. We crossed BAC-TREM2- R47H mice with 5xFAD [1], an amyloid depositing mouse model of AD, to evaluate amyloid pathologies and microglial phenotypes, transcriptomics and in situ expression of key TREM2 -dosage dependent genes. We also compared the key findings in 5xFAD/BAC-TREM2-R47H to those observed in 5xFAD/BAC-TREM2 mice. Result: Both BAC-TREM2 and BAC-TREM2-R47H showed proper expression of three splicing isoforms of TREM2 that are normally found in human. In 5xFAD background, elevated TREM2-R47H gene dosages significantly reduced the plaque burden, especially the filamentous type. The results were consistent with enhanced phagocytosis and altered NLRP3 inflammasome activation in BAC- TREM2-R47H microglia in vitro. However, unlike TREM2 overexpression, elevated TREM2- R47H in 5xFAD failed to ameliorate cognitive and transcriptomic deficits. In situ analysis of key TREM2 -dosage dependent genes and microglial morphology uncovered that TREM2-R47H showed a loss-of-function phenotype in reprogramming of plaque-associated microglial reactivity and gene expression in 5xFAD. Conclusion: Our study demonstrated that the AD-risk variant has a previously unknown, mixture of partial and full loss of TREM2 functions in modulating microglial response in AD mouse brains. Together, our new BAC-TREM2-R47H model and prior BAC-TREM2 mice are invaluable resource to facilitate the therapeutic discovery that target human TREM2 and its R47H variant to ameliorate AD and other neurodegenerative disorders.

2.
Mol Neurodegener ; 17(1): 44, 2022 06 18.
Article in English | MEDLINE | ID: mdl-35717259

ABSTRACT

BACKGROUND: Microglia plays crucial roles in Alzheimer's disease (AD) development. Triggering receptor expressed on myeloid cells 2 (TREM2) in association with DAP12 mediates signaling affecting microglia function. Here we study the negative regulation of TREM2 functions by leukocyte immunoglobulin-like receptor subfamily B member 2 (LILRB2), an inhibitory receptor bearing ITIM motifs. METHODS: To specifically interrogate LILRB2-ligand (oAß and PS) interactions and microglia functions, we generated potent antagonistic LILRB2 antibodies with sub-nanomolar level activities. The biological effects of LILRB2 antagonist antibody (Ab29) were studied in human induced pluripotent stem cell (iPSC)-derived microglia (hMGLs) for migration, oAß phagocytosis, and upregulation of inflammatory cytokines. Effects of the LILRB2 antagonist antibody on microglial responses to amyloid plaques were further studied in vivo using stereotaxic grafted microglia in 5XFAD mice. RESULTS: We confirmed the expression of both LILRB2 and TREM2 in human brain microglia using immunofluorescence. Upon co-ligation of the LILRB2 and TREM2 by shared ligands oAß or PS, TREM2 signaling was significantly inhibited. We identified a monoclonal antibody (Ab29) that blocks LILRB2/ligand interactions and prevents TREM2 signaling inhibition mediated by LILRB2. Further, Ab29 enhanced microglia phagocytosis, TREM2 signaling, migration, and cytokine responses to the oAß-lipoprotein complex in hMGL and microglia cell line HMC3. In vivo studies showed significantly enhanced clustering of microglia around plaques with a prominent increase in microglial amyloid plaque phagocytosis when 5XFAD mice were treated with Ab29. CONCLUSIONS: This study revealed for the first time the molecular mechanisms of LILRB2-mediated inhibition of TREM2 signaling in microglia and demonstrated a novel approach of enhancing TREM2-mediated microglia functions by blocking LILRB2-ligand interactions. Translationally, a LILRB2 antagonist antibody completely rescued the inhibition of TREM2 signaling by LILRB2, suggesting a novel therapeutic strategy for improving microglial functions.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Ligands , Membrane Glycoproteins/metabolism , Mice , Microglia/metabolism , Plaque, Amyloid/metabolism , Receptors, Immunologic/metabolism
3.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34156424

ABSTRACT

Biochemical, pathogenic, and human genetic data confirm that GSAP (γ-secretase activating protein), a selective γ-secretase modulatory protein, plays important roles in Alzheimer's disease (AD) and Down's syndrome. However, the molecular mechanism(s) underlying GSAP-dependent pathogenesis remains largely elusive. Here, through unbiased proteomics and single-nuclei RNAseq, we identified that GSAP regulates multiple biological pathways, including protein phosphorylation, trafficking, lipid metabolism, and mitochondrial function. We demonstrated that GSAP physically interacts with the Fe65-APP complex to regulate APP trafficking/partitioning. GSAP is enriched in the mitochondria-associated membrane (MAM) and regulates lipid homeostasis through the amyloidogenic processing of APP. GSAP deletion generates a lipid environment unfavorable for AD pathogenesis, leading to improved mitochondrial function and the rescue of cognitive deficits in an AD mouse model. Finally, we identified a novel GSAP single-nucleotide polymorphism that regulates its brain transcript level and is associated with an increased AD risk. Together, our findings indicate that GSAP impairs mitochondrial function through its MAM localization and that lowering GSAP expression reduces pathological effects associated with AD.


Subject(s)
Alzheimer Disease/pathology , Homeostasis , Lipid Metabolism , Mitochondria/metabolism , Proteins/metabolism , Aging/pathology , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Base Sequence , Disease Models, Animal , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Membranes/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Open Field Test , Phosphorylation , Protein Binding , Protein Transport , Proteins/genetics , Transcription, Genetic
4.
J Neurosci ; 40(31): 5908-5921, 2020 07 29.
Article in English | MEDLINE | ID: mdl-32601248

ABSTRACT

SORLA is a transmembrane trafficking protein associated with Alzheimer's disease risk. Although SORLA is abundantly expressed in neurons, physiological roles for SORLA remain unclear. Here, we show that cultured transgenic neurons overexpressing SORLA feature longer neurites, and accelerated neurite regeneration with wounding. Enhanced release of a soluble form of SORLA (sSORLA) is observed in transgenic mouse neurons overexpressing human SORLA, while purified sSORLA promotes neurite extension and regeneration. Phosphoproteomic analyses demonstrate enrichment of phosphoproteins related to the epidermal growth factor (EGFR)/ERK pathway in SORLA transgenic mouse hippocampus from both genders. sSORLA coprecipitates with EGFR in vitro, and sSORLA treatment increases EGFR Y1173 phosphorylation, which is involved in ERK activation in cultured neurons. Furthermore, sSORLA triggers ERK activation, whereas pharmacological EGFR or ERK inhibition reverses sSORLA-dependent enhancement of neurite outgrowth. In search for downstream ERK effectors activated by sSORLA, we identified upregulation of Fos expression in hippocampus from male mice overexpressing SORLA by RNAseq analysis. We also found that Fos is upregulated and translocates to the nucleus in an ERK-dependent manner in neurons treated with sSORLA. Together, these results demonstrate that sSORLA is an EGFR-interacting protein that activates EGFR/ERK/Fos signaling to enhance neurite outgrowth and regeneration.SIGNIFICANCE STATEMENT SORLA is a transmembrane trafficking protein previously known to reduce the levels of amyloid-ß, which is critical in the pathogenesis of Alzheimer's disease. In addition, SORLA mutations are a risk factor for Alzheimer's disease. Interestingly, the SORLA ectodomain is cleaved into a soluble form, sSORLA, which has been shown to regulate cytoskeletal signaling pathways and cell motility in cells outside the nervous system. We show here that sSORLA binds and activates the EGF receptor to induce downstream signaling through the ERK serine/threonine kinase and the Fos transcription factor, thereby enhancing neurite outgrowth. These findings reveal a novel role for sSORLA in promoting neurite regeneration through the EGF receptor/ERK/Fos pathway, thereby demonstrating a potential neuroprotective mechanism involving SORLA.


Subject(s)
ErbB Receptors/physiology , MAP Kinase Signaling System/physiology , Membrane Transport Proteins/physiology , Nerve Regeneration/physiology , Neurites/physiology , Receptors, LDL/physiology , Animals , Cells, Cultured , Female , Gene Expression Regulation , Genes, fos , Hippocampus/physiology , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Phosphorylation , Receptors, LDL/genetics
5.
J Clin Invest ; 129(8): 3103-3120, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31112137

ABSTRACT

Mechanisms underlying motor neuron degeneration in amyotrophic lateral sclerosis (ALS) are yet unclear. Specific deletion of the ER-component membralin in astrocytes manifested postnatal motor defects and lethality in mice, causing the accumulation of extracellular glutamate through reducing the glutamate transporter EAAT2. Restoring EAAT2 levels in membralin KO astrocytes limited astrocyte-dependent excitotoxicity in motor neurons. Transcriptomic profiles from mouse astrocytic membralin KO motor cortex indicated significant perturbation in KEGG pathway components related to ALS, including downregulation of Eaat2 and upregulation of Tnfrsf1a. Changes in gene expression with membralin deletion also overlapped with mouse ALS models and reactive astrocytes. Our results shown that activation of TNF receptor (TNFR1)-NFκB pathway known to suppress Eaat2 transcription was upregulated with membralin deletion. Further, reduced membralin and EAAT2 levels correlated with disease progression in spinal cord from SOD1-mutant mouse models, and reductions in membralin/EAAT2 were observed in human ALS spinal cord. Importantly, overexpression of membralin in SOD1G93A astrocytes decreased TNFR1 levels and increased EAAT2 expression, and improved motor neuron survival. Importantly, upregulation of membralin in SOD1G93A mice significantly prolonged mouse survival. Together, our study provided a mechanism for ALS pathogenesis where membralin limited glutamatergic neurotoxicity, suggesting that modulating membralin had potentials in ALS therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Glutamic Acid/metabolism , Motor Cortex/metabolism , Nerve Tissue Proteins/deficiency , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Astrocytes/pathology , Down-Regulation , Excitatory Amino Acid Transporter 2/biosynthesis , Excitatory Amino Acid Transporter 2/genetics , Glutamic Acid/genetics , Humans , Mice , Mice, Knockout , Motor Cortex/pathology , Nerve Tissue Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/genetics , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Transcription, Genetic , Up-Regulation
6.
J Neurosci ; 38(35): 7683-7700, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30054395

ABSTRACT

Aging contributes to cellular stress and neurodegeneration. Our understanding is limited regarding the tissue-restricted mechanisms providing protection in postmitotic cells throughout life. Here, we show that spinal cord motoneurons exhibit a high abundance of asymmetric dimethyl arginines (ADMAs) and the presence of this posttranslational modification provides protection against environmental stress. We identify protein arginine methyltransferase 8 (PRMT8) as a tissue-restricted enzyme responsible for proper ADMA level in postmitotic neurons. Male PRMT8 knock-out mice display decreased muscle strength with aging due to premature destabilization of neuromuscular junctions. Mechanistically, inhibition of methyltransferase activity or loss of PRMT8 results in accumulation of unrepaired DNA double-stranded breaks and decrease in the cAMP response-element-binding protein 1 (CREB1) level. As a consequence, the expression of CREB1-mediated prosurvival and regeneration-associated immediate early genes is dysregulated in aging PRMT8 knock-out mice. The uncovered role of PRMT8 represents a novel mechanism of stress tolerance in long-lived postmitotic neurons and identifies PRMT8 as a tissue-specific therapeutic target in the prevention of motoneuron degeneration.SIGNIFICANCE STATEMENT Although most of the cells in our body have a very short lifespan, postmitotic neurons must survive for many decades. Longevity of a cell within the organism depends on its ability to properly regulate signaling pathways that counteract perturbations, such as DNA damage, oxidative stress, or protein misfolding. Here, we provide evidence that tissue-specific regulators of stress tolerance exist in postmitotic neurons. Specifically, we identify protein arginine methyltransferase 8 (PRMT8) as a cell-type-restricted arginine methyltransferase in spinal cord motoneurons (MNs). PRMT8-dependent arginine methylation is required for neuroprotection against age-related increased of cellular stress. Tissue-restricted expression and the enzymatic activity of PRMT8 make it an attractive target for drug development to delay the onset of neurodegenerative disorders.


Subject(s)
DNA Damage/physiology , Motor Neurons/enzymology , Protein-Arginine N-Methyltransferases/physiology , Aging/metabolism , Amino Acid Sequence , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Cell Line , Cyclic AMP Response Element-Binding Protein/physiology , DNA Breaks, Double-Stranded , DNA Repair , Isometric Contraction , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Cells/enzymology , Muscle Cells/physiology , Neuromuscular Junction/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , RNA Interference , RNA, Small Interfering/pharmacology , Recombinant Fusion Proteins/metabolism , Reflex, Abnormal , Rotarod Performance Test , Spinal Cord/cytology , Spinal Cord/growth & development
7.
Neuron ; 97(5): 1023-1031.e7, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29518356

ABSTRACT

Mutations in triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to increased Alzheimer's disease (AD) risk. Neurobiological functions of TREM2 and its pathophysiological ligands remain elusive. Here we found that TREM2 directly binds to ß-amyloid (Aß) oligomers with nanomolar affinity, whereas AD-associated TREM2 mutations reduce Aß binding. TREM2 deficiency impairs Aß degradation in primary microglial culture and mouse brain. Aß-induced microglial depolarization, K+ inward current induction, cytokine expression and secretion, migration, proliferation, apoptosis, and morphological changes are dependent on TREM2. In addition, TREM2 interaction with its signaling adaptor DAP12 is enhanced by Aß, regulating downstream phosphorylation of SYK and GSK3ß. Our data demonstrate TREM2 as a microglial Aß receptor transducing physiological and AD-related pathological effects associated with Aß.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Membrane Glycoproteins/metabolism , Microglia/physiology , Receptors, Immunologic/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Animals , Brain/pathology , Cells, Cultured , Double-Blind Method , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/pathology , Protein Binding/physiology
8.
Neuron ; 97(5): 1032-1048.e5, 2018 03 07.
Article in English | MEDLINE | ID: mdl-29518357

ABSTRACT

Variants of TREM2 are associated with Alzheimer's disease (AD). To study whether increasing TREM2 gene dosage could modify the disease pathogenesis, we developed BAC transgenic mice expressing human TREM2 (BAC-TREM2) in microglia. We found that elevated TREM2 expression reduced amyloid burden in the 5xFAD mouse model. Transcriptomic profiling demonstrated that increasing TREM2 levels conferred a rescuing effect, which includes dampening the expression of multiple disease-associated microglial genes and augmenting downregulated neuronal genes. Interestingly, 5xFAD/BAC-TREM2 mice showed further upregulation of several reactive microglial genes linked to phagocytosis and negative regulation of immune cell activation. Moreover, these mice showed enhanced process ramification and phagocytic marker expression in plaque-associated microglia and reduced neuritic dystrophy. Finally, elevated TREM2 gene dosage led to improved memory performance in AD models. In summary, our study shows that a genomic transgene-driven increase in TREM2 expression reprograms microglia responsivity and ameliorates neuropathological and behavioral deficits in AD mouse models.


Subject(s)
Alzheimer Disease/genetics , Disease Models, Animal , Gene Dosage/genetics , Membrane Glycoproteins/genetics , Microglia/physiology , Phenotype , Receptors, Immunologic/genetics , Alzheimer Disease/pathology , Animals , Animals, Newborn , Cells, Cultured , Cellular Reprogramming Techniques/methods , Female , Humans , Male , Membrane Glycoproteins/biosynthesis , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , Organ Culture Techniques , Receptors, Immunologic/biosynthesis
9.
J Exp Med ; 214(12): 3669-3685, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-29114064

ABSTRACT

Sortilin-related receptor with LDLR class A repeats (SORLA, SORL1, or LR11) is a genetic risk factor associated with Alzheimer's disease (AD). Although SORLA is known to regulate trafficking of the amyloid ß (Aß) precursor protein to decrease levels of proteotoxic Aß oligomers, whether SORLA can counteract synaptic dysfunction induced by Aß oligomers remains unclear. Here, we show that SORLA interacts with the EphA4 receptor tyrosine kinase and attenuates ephrinA1 ligand-induced EphA4 clustering and activation to limit downstream effects of EphA4 signaling in neurons. Consistent with these findings, SORLA transgenic mice, compared with WT mice, exhibit decreased EphA4 activation and redistribution to postsynaptic densities, with milder deficits in long-term potentiation and memory induced by Aß oligomers. Importantly, we detected elevated levels of active EphA4 in human AD brains, where EphA4 activation is inversely correlated with SORLA/EphA4 association. These results demonstrate a novel role for SORLA as a physiological and pathological EphA4 modulator, which attenuates synaptotoxic EphA4 activation and cognitive impairment associated with Aß-induced neurodegeneration in AD.


Subject(s)
Amyloid beta-Protein Precursor/toxicity , LDL-Receptor Related Proteins/metabolism , Membrane Transport Proteins/metabolism , Nerve Degeneration/pathology , Receptor, EphA4/metabolism , Receptors, LDL/metabolism , Alzheimer Disease/pathology , Animals , Ephrins/pharmacology , Growth Cones/drug effects , Growth Cones/metabolism , HEK293 Cells , Humans , Ligands , Long-Term Potentiation/drug effects , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Mice, Inbred BALB C , Mice, Transgenic , Mutation/genetics , Nerve Degeneration/metabolism , Protein Binding/drug effects , Protein Domains , Receptors, LDL/chemistry , Receptors, LDL/genetics , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
10.
Exp Mol Med ; 49(12): e405, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29611543

ABSTRACT

Genetic mutations in triggering receptor expressed on myeloid cells 2 (TREM2) have been linked to a variety of neurodegenerative diseases including Alzheimer's disease, amyotrophic lateral sclerosis, frontotemporal dementia and Parkinson's disease. In the brain, TREM2 is highly expressed on the cell surface of microglia, where it can transduce signals to regulate microglial functions such as phagocytosis. To date, mechanisms underlying intracellular trafficking of TREM2 remain elusive. Mutations in the presenilin 1 (PS1) catalytic subunit of the γ-secretase complex have been associated with increased generation of the amyloidogenic Aß (amyloid-ß) 42 peptide through cleavage of the Aß precursor amyloid precursor protein. Here we found that TREM2 interacts with PS1 in a manner independent of γ-secretase activity. Mutations in TREM2 alter its subcellular localization and affects its interaction with PS1. Upregulation of PS1 reduces, whereas downregulation of PS1 increases, steady-state levels of cell surface TREM2. Furthermore, PS1 overexpression results in attenuated phagocytic uptake of Aß by microglia, which is reversed by TREM2 overexpression. Our data indicate a novel role for PS1 in regulating TREM2 intracellular trafficking and pathophysiological function.


Subject(s)
Membrane Glycoproteins/metabolism , Presenilin-1/metabolism , Receptors, Immunologic/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Biotinylation , Cell Line , HEK293 Cells , Humans , Immunoprecipitation , Membrane Glycoproteins/genetics , Mice , Mutation/genetics , Phagocytosis/genetics , Phagocytosis/physiology , Presenilin-1/genetics , Protein Transport/genetics , Protein Transport/physiology , Receptors, Immunologic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...