Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 185(4): 712-728.e14, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35063084

ABSTRACT

Tau (MAPT) drives neuronal dysfunction in Alzheimer disease (AD) and other tauopathies. To dissect the underlying mechanisms, we combined an engineered ascorbic acid peroxidase (APEX) approach with quantitative affinity purification mass spectrometry (AP-MS) followed by proximity ligation assay (PLA) to characterize Tau interactomes modified by neuronal activity and mutations that cause frontotemporal dementia (FTD) in human induced pluripotent stem cell (iPSC)-derived neurons. We established interactions of Tau with presynaptic vesicle proteins during activity-dependent Tau secretion and mapped the Tau-binding sites to the cytosolic domains of integral synaptic vesicle proteins. We showed that FTD mutations impair bioenergetics and markedly diminished Tau's interaction with mitochondria proteins, which were downregulated in AD brains of multiple cohorts and correlated with disease severity. These multimodal and dynamic Tau interactomes with exquisite spatial resolution shed light on Tau's role in neuronal function and disease and highlight potential therapeutic targets to block Tau-mediated pathogenesis.


Subject(s)
Mitochondria/metabolism , Nerve Degeneration/metabolism , Protein Interaction Maps , Synapses/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Amino Acids/metabolism , Biotinylation , Brain/metabolism , Brain/pathology , Cell Nucleus/metabolism , Disease Progression , Energy Metabolism , Frontotemporal Dementia/genetics , Humans , Induced Pluripotent Stem Cells/metabolism , Mutant Proteins/metabolism , Mutation/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Protein Binding , Protein Domains , Proteomics , Severity of Illness Index , Subcellular Fractions/metabolism , Tauopathies/genetics , tau Proteins/chemistry
2.
Proc Natl Acad Sci U S A ; 121(31): e2323050121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39042684

ABSTRACT

Cerebellar injury in preterm infants with central nervous system (CNS) hemorrhage results in lasting neurological deficits and an increased risk of autism. The impact of blood-induced pathways on cerebellar development remains largely unknown, so no specific treatments have been developed to counteract the harmful effects of blood after neurovascular damage in preterm infants. Here, we show that fibrinogen, a blood-clotting protein, plays a central role in impairing neonatal cerebellar development. Longitudinal MRI of preterm infants revealed that cerebellar bleeds were the most critical factor associated with poor cerebellar growth. Using inflammatory and hemorrhagic mouse models of neonatal cerebellar injury, we found that fibrinogen increased innate immune activation and impeded neurogenesis in the developing cerebellum. Fibrinogen inhibited sonic hedgehog (SHH) signaling, the main mitogenic pathway in cerebellar granule neuron progenitors (CGNPs), and was sufficient to disrupt cerebellar growth. Genetic fibrinogen depletion attenuated neuroinflammation, promoted CGNP proliferation, and preserved normal cerebellar development after neurovascular damage. Our findings suggest that fibrinogen alters the balance of SHH signaling in the neurovascular niche and may serve as a therapeutic target to mitigate developmental brain injury after CNS hemorrhage.


Subject(s)
Blood-Brain Barrier , Cerebellum , Fibrinogen , Hedgehog Proteins , Signal Transduction , Hedgehog Proteins/metabolism , Animals , Fibrinogen/metabolism , Cerebellum/metabolism , Mice , Blood-Brain Barrier/metabolism , Humans , Animals, Newborn , Infant, Newborn , Neurogenesis , Female , Male , Disease Models, Animal
3.
PLoS Biol ; 17(2): e3000134, 2019 02.
Article in English | MEDLINE | ID: mdl-30735499

ABSTRACT

Microglia are resident immune cells that play critical roles in maintaining the normal physiology of the central nervous system (CNS). Remarkably, microglia have an intrinsic capacity to repopulate themselves after acute ablation. However, the underlying mechanisms that drive such restoration remain elusive. Here, we characterized microglial repopulation both spatially and temporally following removal via treatment with the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX5622. We show that microglia were replenished via self-renewal, with no contribution from nonmicroglial lineages, including Nestin+ progenitors and the circulating myeloid population. Interestingly, spatial analyses with dual-color labeling revealed that newborn microglia recolonized the parenchyma by forming distinctive clusters that maintained stable territorial boundaries over time, indicating the proximal expansive nature of adult microgliogenesis and the stability of microglia tiling. Temporal transcriptome profiling at different repopulation stages revealed that adult newborn microglia gradually regain steady-state maturity from an immature state that is reminiscent of the neonatal stage and follow a series of maturation programs, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation, interferon immune activation, and apoptosis. Importantly, we show that the restoration of microglial homeostatic density requires NF-κB signaling as well as apoptotic egress of excessive cells. In summary, our study reports key events that take place from microgliogenesis to homeostasis reestablishment.


Subject(s)
Aging/genetics , Brain/metabolism , Homeostasis/genetics , Microglia/metabolism , NF-kappa B/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Aging/metabolism , Animals , Animals, Newborn , Apoptosis/drug effects , Apoptosis/genetics , Brain/cytology , Brain/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Developmental , Interferons/genetics , Interferons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/cytology , Microglia/drug effects , NF-kappa B/metabolism , Nestin/genetics , Nestin/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Organic Chemicals/toxicity , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Platelet-Derived Growth Factor/genetics , Receptors, Platelet-Derived Growth Factor/metabolism , Regeneration/genetics , Signal Transduction , Transcriptome
4.
Brain ; 144(8): 2291-2301, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34426831

ABSTRACT

Extrinsic inhibitors at sites of blood-brain barrier disruption and neurovascular damage contribute to remyelination failure in neurological diseases. However, therapies to overcome the extrinsic inhibition of remyelination are not widely available and the dynamics of glial progenitor niche remodelling at sites of neurovascular dysfunction are largely unknown. By integrating in vivo two-photon imaging co-registered with electron microscopy and transcriptomics in chronic neuroinflammatory lesions, we found that oligodendrocyte precursor cells clustered perivascularly at sites of limited remyelination with deposition of fibrinogen, a blood coagulation factor abundantly deposited in multiple sclerosis lesions. By developing a screen (OPC-X-screen) to identify compounds that promote remyelination in the presence of extrinsic inhibitors, we showed that known promyelinating drugs did not rescue the extrinsic inhibition of remyelination by fibrinogen. In contrast, bone morphogenetic protein type I receptor blockade rescued the inhibitory fibrinogen effects and restored a promyelinating progenitor niche by promoting myelinating oligodendrocytes, while suppressing astrocyte cell fate, with potent therapeutic effects in chronic models of multiple sclerosis. Thus, abortive oligodendrocyte precursor cell differentiation by fibrinogen is refractory to known promyelinating compounds, suggesting that blockade of the bone morphogenetic protein signalling pathway may enhance remyelinating efficacy by overcoming extrinsic inhibition in neuroinflammatory lesions with vascular damage.


Subject(s)
Blood-Brain Barrier/drug effects , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Oligodendroglia/drug effects , Remyelination/drug effects , Spinal Cord/drug effects , Animals , Blood-Brain Barrier/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Homeostasis/drug effects , Mice , Mice, Transgenic , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/drug effects , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , Spinal Cord/metabolism
5.
Proc Natl Acad Sci U S A ; 115(40): 10172-10177, 2018 10 02.
Article in English | MEDLINE | ID: mdl-30232263

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia, is characterized by the abnormal accumulation of amyloid plaques and hyperphosphorylated tau aggregates, as well as microgliosis. Hemizygous missense variants in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) are associated with elevated risk for developing late-onset AD. These variants are hypothesized to result in loss of function, mimicking TREM2 haploinsufficiency. However, the consequences of TREM2 haploinsufficiency on tau pathology and microglial function remain unknown. We report the effects of partial and complete loss of TREM2 on microglial function and tau-associated deficits. In vivo imaging revealed that microglia from aged TREM2-haploinsufficient mice show a greater impairment in their injury response compared with microglia from aged TREM2-KO mice. In transgenic mice expressing mutant human tau, TREM2 haploinsufficiency, but not complete loss of TREM2, increased tau pathology. In addition, whereas complete TREM2 deficiency protected against tau-mediated microglial activation and atrophy, TREM2 haploinsufficiency elevated expression of proinflammatory markers and exacerbated atrophy at a late stage of disease. The differential effects of partial and complete loss of TREM2 on microglial function and tau pathology provide important insights into the critical role of TREM2 in AD pathogenesis.


Subject(s)
Alzheimer Disease , Haploinsufficiency , Hemizygote , Membrane Glycoproteins , Microglia/metabolism , Mutation, Missense , Receptors, Immunologic , Aging/genetics , Aging/metabolism , Aging/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microglia/pathology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism
6.
J Neurosci ; 35(2): 807-18, 2015 Jan 14.
Article in English | MEDLINE | ID: mdl-25589773

ABSTRACT

Aging is the predominant risk factor for neurodegenerative diseases. One key phenotype as the brain ages is an aberrant innate immune response characterized by proinflammation. However, the molecular mechanisms underlying aging-associated proinflammation are poorly defined. Whether chronic inflammation plays a causal role in cognitive decline in aging and neurodegeneration has not been established. Here we report a mechanistic link between chronic inflammation and aging microglia and a causal role of aging microglia in neurodegenerative cognitive deficits. We showed that SIRT1 is reduced with the aging of microglia and that microglial SIRT1 deficiency has a causative role in aging- or tau-mediated memory deficits via IL-1ß upregulation in mice. Interestingly, the selective activation of IL-1ß transcription by SIRT1 deficiency is likely mediated through hypomethylating the specific CpG sites on IL-1ß proximal promoter. In humans, hypomethylation of IL-1ß is strongly associated with chronological age and with elevated IL-1ß transcription. Our findings reveal a novel epigenetic mechanism in aging microglia that contributes to cognitive deficits in aging and neurodegenerative diseases.


Subject(s)
Aging/metabolism , Cognition , Epigenesis, Genetic , Interleukin-1beta/metabolism , Microglia/metabolism , Sirtuin 1/metabolism , Animals , Case-Control Studies , DNA Methylation , Humans , Interleukin-1beta/genetics , Mice , Sirtuin 1/deficiency , Sirtuin 1/genetics , Tauopathies/metabolism , Up-Regulation
7.
J Biol Chem ; 287(47): 39834-41, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23024364

ABSTRACT

Accumulation of amyloid-ß (Aß), believed to be a key trigger of Alzheimer disease (AD), could result from impaired clearance mechanisms. Previously, we showed that the cysteine protease cathepsin B (CatB) degrades Aß, most likely by C-terminal truncation, in mice expressing human amyloid precursor protein with familial AD-linked mutations (hAPP(FAD)). In addition, the Aß-degrading activity of CatB is inhibited by its endogenous inhibitor, cystatin C (CysC). Reducing CysC expression markedly lowers Aß levels by enhancing CatB-mediated Aß degradation in hAPP(FAD) mice. However, because a vast majority of AD patients do not carry familial mutations, we investigated how the CysC-CatB axis affects Aß levels in mice expressing wild-type hAPP (hAPP(WT)). Enhancing CatB activity by CysC deletion significantly lowered total Aß and Aß42 levels in hAPP(WT) mice, whereas CatB deletion increased Aß levels. To determine whether neuron-derived CatB degrades Aß in vivo, we generated transgenic mice overexpressing CatB under the control of a neuron-specific enolase promoter. Enhancing neuronal CatB activity in hAPP(WT) mice significantly lowered Aß42 levels. The processing of hAPP(WT) was unaffected by increasing or ablating CatB activity. Thus, the CysC-CatB axis affects degradation of Aß42 derived from hAPP lacking familial mutations. These findings support the notion that enhancing CatB activity could lower Aß, especially Aß42, in AD patients with or without familial mutations.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Cathepsin B/metabolism , Genetic Diseases, Inborn/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Proteolysis , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Cathepsin B/genetics , Cystatins/genetics , Cystatins/metabolism , Gene Deletion , Gene Expression , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Humans , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Neurons/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism
8.
J Biol Chem ; 286(37): 32713-22, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21771791

ABSTRACT

Aberrant microglial activation has been proposed to contribute to the cognitive decline in Alzheimer disease (AD), but the underlying molecular mechanisms remain enigmatic. Fractalkine signaling, a pathway mediating the communication between microglia and neurons, is deficient in AD brains and down-regulated by amyloid-ß. Although fractalkine receptor (CX3CR1) on microglia was found to regulate plaque load, no functional effects have been reported. Our study demonstrates that CX3CR1 deficiency worsens the AD-related neuronal and behavioral deficits. The effects were associated with cytokine production but not with plaque deposition. Ablation of CX3CR1 in mice overexpressing human amyloid precursor protein enhanced Tau pathology and exacerbated the depletion of calbindin in the dentate gyrus. The levels of calbindin in the dentate gyrus correlated negatively with those of tumor necrosis factor α and interleukin 6, suggesting neurotoxic effects of inflammatory factors. Functionally, removing CX3CR1 in human amyloid precursor protein mice worsened the memory retention in passive avoidance and novel object recognition tests, and their memory loss in the novel object recognition test is associated with high levels of interleukin 6. Our findings identify CX3CR1 as a key microglial pathway in protecting against AD-related cognitive deficits that are associated with aberrant microglial activation and elevated inflammatory cytokines.


Subject(s)
Alzheimer Disease/metabolism , Cognition Disorders/metabolism , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , CX3C Chemokine Receptor 1 , Calbindins , Cognition Disorders/genetics , Cognition Disorders/pathology , Cytokines/genetics , Cytokines/metabolism , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Disease Models, Animal , Humans , Mice , Mice, Knockout , Microglia/pathology , Nerve Tissue Proteins/genetics , Receptors, Chemokine/genetics , S100 Calcium Binding Protein G/genetics , S100 Calcium Binding Protein G/metabolism
9.
J Neuroinflammation ; 9: 22, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22272874

ABSTRACT

BACKGROUND: Microglia, the resident immune cells of the brain, have been implicated in brain injury and various neurological disorders. However, their precise roles in different pathophysiological situations remain enigmatic and may range from detrimental to protective. Targeting the delivery of biologically active compounds to microglia could help elucidate these roles and facilitate the therapeutic modulation of microglial functions in neurological diseases. METHODS: Here we employ primary cell cultures and stereotaxic injections into mouse brain to investigate the cell type specific localization of semiconductor quantum dots (QDs) in vitro and in vivo. Two potential receptors for QDs are identified using pharmacological inhibitors and neutralizing antibodies. RESULTS: In mixed primary cortical cultures, QDs were selectively taken up by microglia; this uptake was decreased by inhibitors of clathrin-dependent endocytosis, implicating the endosomal pathway as the major route of entry for QDs into microglia. Furthermore, inhibiting mannose receptors and macrophage scavenger receptors blocked the uptake of QDs by microglia, indicating that QD uptake occurs through microglia-specific receptor endocytosis. When injected into the brain, QDs were taken up primarily by microglia and with high efficiency. In primary cortical cultures, QDs conjugated to the toxin saporin depleted microglia in mixed primary cortical cultures, protecting neurons in these cultures against amyloid beta-induced neurotoxicity. CONCLUSIONS: These findings demonstrate that QDs can be used to specifically label and modulate microglia in primary cortical cultures and in brain and may allow for the selective delivery of therapeutic agents to these cells.


Subject(s)
Brain/cytology , Microglia/physiology , Quantum Dots , Amyloid beta-Peptides/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , CX3C Chemokine Receptor 1 , Calcium-Binding Proteins/metabolism , Cell Death/drug effects , Cerebral Cortex/cytology , Clathrin/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Endocytosis/drug effects , Endocytosis/physiology , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Immunotoxins/pharmacology , Mannans/pharmacology , Mice , Mice, Transgenic , Microfilament Proteins/metabolism , Microglia/drug effects , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/physiology , Peptide Fragments/pharmacology , Poly I/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/genetics , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Stereotaxic Techniques , Time Factors
10.
Nat Commun ; 13(1): 1969, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35413950

ABSTRACT

Activation of microglia is a prominent pathological feature in tauopathies, including Alzheimer's disease. How microglia activation contributes to tau toxicity remains largely unknown. Here we show that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, activated by tau, drives microglial-mediated tau propagation and toxicity. Constitutive activation of microglial NF-κB exacerbated, while inactivation diminished, tau seeding and spreading in young PS19 mice. Inhibition of NF-κB activation enhanced the retention while reduced the release of internalized pathogenic tau fibrils from primary microglia and rescued microglial autophagy deficits. Inhibition of microglial NF-κB in aged PS19 mice rescued tau-mediated learning and memory deficits, restored overall transcriptomic changes while increasing neuronal tau inclusions. Single cell RNA-seq revealed that tau-associated disease states in microglia were diminished by NF-κB inactivation and further transformed by constitutive NF-κB activation. Our study establishes a role for microglial NF-κB signaling in mediating tau spreading and toxicity in tauopathy.


Subject(s)
Microglia , NF-kappa B , Tauopathies , tau Proteins , Animals , Mice , Microglia/metabolism , Microglia/pathology , NF-kappa B/metabolism , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/metabolism
11.
Sci Transl Med ; 13(622): eabe3947, 2021 12.
Article in English | MEDLINE | ID: mdl-34851693

ABSTRACT

The hemizygous R47H variant of triggering receptor expressed on myeloid cells 2 (TREM2), a microglia-specific gene in the brain, increases risk for late-onset Alzheimer's disease (AD). Using transcriptomic analysis of single nuclei from brain tissues of patients with AD carrying the R47H mutation or the common variant (CV)­TREM2, we found that R47H-associated microglial subpopulations had enhanced inflammatory signatures reminiscent of previously identified disease-associated microglia (DAM) and hyperactivation of AKT, one of the signaling pathways downstream of TREM2. We established a tauopathy mouse model with heterozygous knock-in of the human TREM2 with the R47H mutation or CV and found that R47H induced and exacerbated TAU-mediated spatial memory deficits in female mice. Single-cell transcriptomic analysis of microglia from these mice also revealed transcriptomic changes induced by R47H that had substantial overlaps with R47H microglia in human AD brains, including robust increases in proinflammatory cytokines, activation of AKT signaling, and elevation of a subset of DAM signatures. Pharmacological AKT inhibition with MK-2206 largely reversed the enhanced inflammatory signatures in primary R47H microglia treated with TAU fibrils. In R47H heterozygous tauopathy mice, MK-2206 treatment abolished a tauopathy-dependent microglial subcluster and rescued tauopathy-induced synapse loss. By uncovering disease-enhancing mechanisms of the R47H mutation conserved in human and mouse, our study supports inhibitors of AKT signaling as a microglial modulating strategy to treat AD.


Subject(s)
Alzheimer Disease , Microglia , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Brain/metabolism , Female , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Microglia/metabolism , Mutation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Immunologic/metabolism
12.
Neuron ; 51(6): 703-14, 2006 Sep 21.
Article in English | MEDLINE | ID: mdl-16982417

ABSTRACT

Alzheimer's disease (AD) may result from the accumulation of amyloid-beta (Abeta) peptides in the brain. The cysteine protease cathepsin B (CatB) is associated with amyloid plaques in AD brains and has been suspected to increase Abeta production. Here, we demonstrate that CatB actually reduces levels of Abeta peptides, especially the aggregation-prone species Abeta1-42, through proteolytic cleavage. Genetic inactivation of CatB in mice with neuronal expression of familial AD-mutant human amyloid precursor protein (hAPP) increased the relative abundance of Abeta1-42, worsening plaque deposition and other AD-related pathologies. Lentivirus-mediated expression of CatB in aged hAPP mice reduced preexisting amyloid deposits, even thioflavin S-positive plaques. Under cell-free conditions, CatB effectively cleaved Abeta1-42, generating C-terminally truncated Abeta peptides that are less amyloidogenic. Thus, CatB likely fulfills antiamyloidogenic and neuroprotective functions. Insufficient CatB activity might promote AD; increasing CatB activity could counteract the neuropathology of this disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Cathepsin B/metabolism , Age Factors , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/ultrastructure , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/metabolism , Brain/pathology , Cathepsin B/genetics , Cell Line, Tumor , Female , Humans , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , Mutation/genetics , Neurites/metabolism , Neurites/pathology , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Peptide Fragments/ultrastructure , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology
13.
Elife ; 92020 10 15.
Article in English | MEDLINE | ID: mdl-33054973

ABSTRACT

Microglia are the resident myeloid cells in the central nervous system (CNS). The majority of microglia rely on CSF1R signaling for survival. However, a small subset of microglia in mouse brains can survive without CSF1R signaling and reestablish the microglial homeostatic population after CSF1R signaling returns. Using single-cell transcriptomic analysis, we characterized the heterogeneous microglial populations under CSF1R inhibition, including microglia with reduced homeostatic markers and elevated markers of inflammatory chemokines and proliferation. Importantly, MAC2/Lgals3 was upregulated under CSF1R inhibition, and shared striking similarities with microglial progenitors in the yolk sac and immature microglia in early embryos. Lineage-tracing studies revealed that these MAC2+ cells were of microglial origin. MAC2+ microglia were also present in non-treated adult mouse brains and exhibited immature transcriptomic signatures indistinguishable from those that survived CSF1R inhibition, supporting the notion that MAC2+ progenitor-like cells are present among adult microglia.


Subject(s)
Brain/metabolism , Galectin 3/genetics , Mice/physiology , Microglia/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Signal Transduction/genetics , Animals , Female , Galectin 3/metabolism , Homeostasis , Male , Mice/genetics , Mice, Inbred C57BL , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
14.
Sci Rep ; 10(1): 13688, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792571

ABSTRACT

Patients with frontotemporal dementia (FTD) resulting from granulin (GRN) haploinsufficiency have reduced levels of progranulin and exhibit dysregulation in inflammatory and lysosomal networks. Microglia produce high levels of progranulin, and reduction of progranulin in microglia alone is sufficient to recapitulate inflammation, lysosomal dysfunction, and hyperproliferation in a cell-autonomous manner. Therefore, targeting microglial dysfunction caused by progranulin insufficiency represents a potential therapeutic strategy to manage neurodegeneration in FTD. Limitations of current progranulin-enhancing strategies necessitate the discovery of new targets. To identify compounds that can reverse microglial defects in Grn-deficient mouse microglia, we performed a compound screen coupled with high throughput sequencing to assess key transcriptional changes in inflammatory and lysosomal pathways. Positive hits from this initial screen were then further narrowed down based on their ability to rescue cathepsin activity, a critical biochemical readout of lysosomal capacity. The screen identified nor-binaltorphimine dihydrochloride (nor-BNI) and dibutyryl-cAMP, sodium salt (DB-cAMP) as two phenotypic modulators of progranulin deficiency. In addition, nor-BNI and DB-cAMP also rescued cell cycle abnormalities in progranulin-deficient cells. These data highlight the potential of a transcription-based platform for drug screening, and advance two novel lead compounds for FTD.


Subject(s)
Bucladesine/pharmacology , Cysteine Proteases/metabolism , Frontotemporal Dementia/genetics , Gene Expression Profiling/methods , Microglia/cytology , Naltrexone/analogs & derivatives , Progranulins/deficiency , Animals , Cell Cycle/drug effects , Cells, Cultured , Disease Models, Animal , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/metabolism , Gene Expression Regulation/drug effects , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing , Humans , Lysosomes/genetics , Lysosomes/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Models, Biological , Naltrexone/pharmacology , Sequence Analysis, RNA , Small Molecule Libraries/pharmacology
15.
Nat Neurosci ; 23(2): 167-171, 2020 02.
Article in English | MEDLINE | ID: mdl-31873194

ABSTRACT

Sex is a key modifier of neurological disease outcomes. Microglia are implicated in neurological diseases and modulated by microRNAs, but it is unknown whether microglial microRNAs have sex-specific influences on disease. We show in mice that microglial microRNA expression differs in males and females and that loss of microRNAs leads to sex-specific changes in the microglial transcriptome and tau pathology. These findings suggest that microglial microRNAs influence tau pathogenesis in a sex-specific manner.


Subject(s)
Brain/pathology , MicroRNAs/metabolism , Microglia/metabolism , Sex Characteristics , Tauopathies/pathology , Animals , Brain/metabolism , Female , Male , Mice , Microglia/pathology , Tauopathies/metabolism , Transcriptome , tau Proteins/metabolism
16.
Alzheimers Dement (N Y) ; 3(4): 507-512, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29124108

ABSTRACT

INTRODUCTION: Frontotemporal lobar degeneration-causing mutations in the progranulin (GRN) gene reduce progranulin protein (PGRN) levels, suggesting that restoring PGRN in mutation carriers may be therapeutic. Nimodipine, a Food and Drug Administration-approved blood-brain barrier-penetrant calcium channel blocker, increased PGRN levels in PGRN-deficient murine models. We sought to assess safety and tolerability of oral nimodipine in human GRN mutation carriers. METHODS: We performed an open-label, 8-week, dose-finding, phase 1 clinical trial in eight GRN mutation carriers to assess the safety and tolerability of nimodipine and assayed fluid and radiologic markers to investigate therapeutic endpoints. RESULTS: There were no serious adverse events; however, PGRN concentrations (cerebrospinal fluid and plasma) did not change significantly following treatment (percent changes of -5.2 ± 10.9% in plasma and -10.2 ± 7.8% in cerebrospinal fluid). Measurable atrophy within the left middle frontal gyrus was observed over an 8-week period. DISCUSSION: While well tolerated, nimodipine treatment did not alter PGRN concentrations or secondary outcomes.

17.
Mol Neurodegener ; 11(1): 47, 2016 06 29.
Article in English | MEDLINE | ID: mdl-27356871

ABSTRACT

BACKGROUND: Neurons are highly polarized cells in which asymmetric axonal-dendritic distribution of proteins is crucial for neuronal function. Loss of polarized distribution of the axonal protein tau is an early sign of Alzheimer's disease (AD) and other neurodegenerative disorders. The cytoskeletal network in the axon initial segment (AIS) forms a barrier between the axon and the somatodentritic compartment, contributing to axonal retention of tau. Although perturbation of the AIS cytoskeleton has been implicated in neurological disorders, the molecular triggers and functional consequence of AIS perturbation are incompletely understood. RESULTS: Here we report that tau acetylation and consequent destabilization of the AIS cytoskeleton promote the somatodendritic mislocalization of tau. AIS cytoskeletal proteins, including ankyrin G and ßIV-spectrin, were downregulated in AD brains and negatively correlated with an increase in tau acetylated at K274 and K281. AIS proteins were also diminished in transgenic mice expressing tauK274/281Q, a tau mutant that mimics K274 and K281 acetylation. In primary neuronal cultures, the tauK274/281Q mutant caused hyperdynamic microtubules (MTs) in the AIS, shown by live-imaging of MT mobility and fluorescence recovery after photobleaching. Using photoconvertible tau constructs, we found that axonal tauK274/281Q was missorted into the somatodendritic compartment. Stabilizing MTs with epothilone D to restore the cytoskeletal barrier in the AIS prevented tau mislocalization in primary neuronal cultures. CONCLUSIONS: Together, these findings demonstrate that tau acetylation contributes to the pathogenesis of neurodegenerative disease by compromising the cytoskeletal sorting machinery in the AIS.


Subject(s)
Alzheimer Disease/pathology , Axon Initial Segment/metabolism , Cell Polarity , Cytoskeleton/pathology , tau Proteins/metabolism , Acetylation , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Animals , Axon Initial Segment/pathology , Blotting, Western , Cell Polarity/physiology , Cytoskeleton/metabolism , Disease Models, Animal , Female , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Neurons/metabolism , Neurons/pathology , Rats
18.
Neuron ; 90(2): 245-60, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27041503

ABSTRACT

Tau toxicity has been implicated in the emergence of synaptic dysfunction in Alzheimer's disease (AD), but the mechanism by which tau alters synapse physiology and leads to cognitive decline is unclear. Here we report abnormal acetylation of K274 and K281 on tau, identified in AD brains, promotes memory loss and disrupts synaptic plasticity by reducing postsynaptic KIdney/BRAin (KIBRA) protein, a memory-associated protein. Transgenic mice expressing human tau with lysine-to-glutamine mutations to mimic K274 and K281 acetylation (tauKQ) exhibit AD-related memory deficits and impaired hippocampal long-term potentiation (LTP). TauKQ reduces synaptic KIBRA levels and disrupts activity-induced postsynaptic actin remodeling and AMPA receptor insertion. The LTP deficit was rescued by promoting actin polymerization or by KIBRA expression. In AD patients with dementia, we found enhanced tau acetylation is linked to loss of KIBRA. These findings suggest a novel mechanism by which pathogenic tau causes synaptic dysfunction and cognitive decline in AD pathogenesis.


Subject(s)
Actins/metabolism , Brain/metabolism , Carrier Proteins/metabolism , Memory Disorders/physiopathology , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Signal Transduction , tau Proteins/metabolism , Acetylation , Alzheimer Disease/metabolism , Animals , Hippocampus/physiology , Humans , Intracellular Signaling Peptides and Proteins , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Memory Disorders/genetics , Mice , Mice, Transgenic , Phosphoproteins , Primary Cell Culture , tau Proteins/genetics
19.
Nat Med ; 21(10): 1154-62, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26390242

ABSTRACT

Tauopathies, including frontotemporal dementia (FTD) and Alzheimer's disease (AD), are neurodegenerative diseases in which tau fibrils accumulate. Recent evidence supports soluble tau species as the major toxic species. How soluble tau accumulates and causes neurodegeneration remains unclear. Here we identify tau acetylation at Lys174 (K174) as an early change in AD brains and a critical determinant in tau homeostasis and toxicity in mice. The acetyl-mimicking mutant K174Q slows tau turnover and induces cognitive deficits in vivo. Acetyltransferase p300-induced tau acetylation is inhibited by salsalate and salicylate, which enhance tau turnover and reduce tau levels. In the PS19 transgenic mouse model of FTD, administration of salsalate after disease onset inhibited p300 activity, lowered levels of total tau and tau acetylated at K174, rescued tau-induced memory deficits and prevented hippocampal atrophy. The tau-lowering and protective effects of salsalate were diminished in neurons expressing K174Q tau. Targeting tau acetylation could be a new therapeutic strategy against human tauopathies.


Subject(s)
Cognition Disorders/physiopathology , Neurodegenerative Diseases/physiopathology , tau Proteins/physiology , Acetylation , Animals , Behavior, Animal , Humans , Mice , tau Proteins/metabolism
20.
Nat Med ; 20(10): 1157-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25261995

ABSTRACT

Haploinsufficiency of the progranulin (PGRN) gene (GRN) causes familial frontotemporal lobar degeneration (FTLD) and modulates an innate immune response in humans and in mouse models. GRN polymorphism may be linked to late-onset Alzheimer's disease (AD). However, the role of PGRN in AD pathogenesis is unknown. Here we show that PGRN inhibits amyloid ß (Aß) deposition. Selectively reducing microglial expression of PGRN in AD mouse models impaired phagocytosis, increased plaque load threefold and exacerbated cognitive deficits. Lentivirus-mediated PGRN overexpression lowered plaque load in AD mice with aggressive amyloid plaque pathology. Aß plaque load correlated negatively with levels of hippocampal PGRN, showing the dose-dependent inhibitory effects of PGRN on plaque deposition. PGRN also protected against Aß toxicity. Lentivirus-mediated PGRN overexpression prevented spatial memory deficits and hippocampal neuronal loss in AD mice. The protective effects of PGRN against Aß deposition and toxicity have important therapeutic implications. We propose enhancing PGRN as a potential treatment for PGRN-deficient FTLD and AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/therapy , Amyloid beta-Peptides/genetics , Animals , Brain/metabolism , Brain/pathology , Cognition/physiology , Disease Models, Animal , Female , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/metabolism , Frontotemporal Lobar Degeneration/therapy , Gene Expression Regulation , Granulins , Humans , Immunity, Innate/physiology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Phagocytosis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Progranulins , Rats , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL