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










Publication year range
1.
Nat Aging ; 3(10): 1219-1236, 2023 10.
Article in English | MEDLINE | ID: mdl-37735240

ABSTRACT

In Alzheimer's disease (AD), sensome receptor dysfunction impairs microglial danger-associated molecular pattern (DAMP) clearance and exacerbates disease pathology. Although extrinsic signals, including interleukin-33 (IL-33), can restore microglial DAMP clearance, it remains largely unclear how the sensome receptor is regulated and interacts with DAMP during phagocytic clearance. Here, we show that IL-33 induces VCAM1 in microglia, which promotes microglial chemotaxis toward amyloid-beta (Aß) plaque-associated ApoE, and leads to Aß clearance. We show that IL-33 stimulates a chemotactic state in microglia, characterized by Aß-directed migration. Functional screening identified that VCAM1 directs microglial Aß chemotaxis by sensing Aß plaque-associated ApoE. Moreover, we found that disrupting VCAM1-ApoE interaction abolishes microglial Aß chemotaxis, resulting in decreased microglial clearance of Aß. In patients with AD, higher cerebrospinal fluid levels of soluble VCAM1 were correlated with impaired microglial Aß chemotaxis. Together, our findings demonstrate that promoting VCAM1-ApoE-dependent microglial functions ameliorates AD pathology.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Microglia/metabolism , Interleukin-33/metabolism , Chemotaxis , Amyloid beta-Peptides/metabolism , Apolipoproteins E/metabolism
2.
J Neurochem ; 166(6): 891-903, 2023 09.
Article in English | MEDLINE | ID: mdl-37603311

ABSTRACT

Microglia maintain brain homeostasis through their ability to survey and phagocytose danger-associated molecular patterns (DAMPs). In Alzheimer's disease (AD), microglial phagocytic clearance regulates the turnover of neurotoxic DAMPs including amyloid beta (Aß) and hyperphosphorylated tau. To mediate DAMP clearance, microglia express a repertoire of surface receptors to sense DAMPs; the activation of these receptors subsequently triggers a chemotaxis-to-phagocytosis functional transition in microglia. Therefore, the interaction between microglial receptors and DAMPs plays a critical role in controlling microglial DAMP clearance and AD pathogenesis. However, there is no comprehensive overview on how microglial sensome receptors interact with DAMPs and regulate various microglial functions, including chemotaxis and phagocytosis. In this review, we discuss the important axes of receptor-ligand interaction that control different microglial functions and their roles in AD pathogenesis. First, we summarize how the accumulation and structural changes of DAMPs trigger microglial functional impairment, including impaired DAMP clearance and aberrant synaptic pruning, in AD. Then, we discuss the important receptor-ligand axes that restore microglial DAMP clearance in AD and aging. These findings suggest that targeting microglial chemotaxis-the first critical step of the microglial chemotaxis-to-phagocytosis state transition-can promote microglial DAMP clearance in AD. Thus, our review highlights the importance of microglial chemotaxis in promoting microglial clearance activity in AD. Further detailed investigations are essential to identify the molecular machinery that controls microglial chemotaxis in AD.


Subject(s)
Alzheimer Disease , Humans , Microglia , Amyloid beta-Peptides , Chemotaxis , Ligands
3.
Nat Aging ; 2(7): 616-634, 2022 07.
Article in English | MEDLINE | ID: mdl-37117777

ABSTRACT

Changes in the levels of circulating proteins are associated with Alzheimer's disease (AD), whereas their pathogenic roles in AD are unclear. Here, we identified soluble ST2 (sST2), a decoy receptor of interleukin-33-ST2 signaling, as a new disease-causing factor in AD. Increased circulating sST2 level is associated with more severe pathological changes in female individuals with AD. Genome-wide association analysis and CRISPR-Cas9 genome editing identified rs1921622 , a genetic variant in an enhancer element of IL1RL1, which downregulates gene and protein levels of sST2. Mendelian randomization analysis using genetic variants, including rs1921622 , demonstrated that decreased sST2 levels lower AD risk and related endophenotypes in females carrying the Apolipoprotein E (APOE)-ε4 genotype; the association is stronger in Chinese than in European-descent populations. Human and mouse transcriptome and immunohistochemical studies showed that rs1921622 /sST2 regulates amyloid-beta (Aß) pathology through the modulation of microglial activation and Aß clearance. These findings demonstrate how sST2 level is modulated by a genetic variation and plays a disease-causing role in females with AD.


Subject(s)
Alzheimer Disease , Humans , Female , Animals , Mice , Alzheimer Disease/genetics , Interleukin-1 Receptor-Like 1 Protein/genetics , Genome-Wide Association Study , Apolipoprotein E4/genetics , Amyloid beta-Peptides/genetics
4.
ACS Chem Neurosci ; 12(22): 4249-4256, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34738783

ABSTRACT

Alzheimer's disease (AD), the most common neurodegenerative disease, has limited treatment options. As such, extensive studies have been conducted to identify novel therapeutic approaches. We previously reported that rhynchophylline (Rhy), a small molecule EphA4 inhibitor, rescues impaired hippocampal synaptic plasticity and cognitive dysfunctions in APP/PS1 mice, an AD transgenic mouse model. To assess whether Rhy can be developed as an alternative treatment for AD, it is important to examine its pharmacokinetics and effects on other disease-associated pathologies. Here, we show that Rhy ameliorates amyloid plaque burden and reduces inflammation in APP/PS1 mice. Transcriptome analysis revealed that Rhy regulates various molecular pathways in APP/PS1 mouse brains associated with amyloid metabolism and inflammation, specifically the ubiquitin proteasome system, angiogenesis, and microglial functional states. These results show that Rhy, which is blood-brain barrier permeable, is beneficial to amyloid pathology and regulates multiple molecular pathways.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Amyloid beta-Protein Precursor/genetics , Animals , Disease Models, Animal , Inflammation/drug therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxindoles , Plaque, Amyloid/drug therapy , Presenilin-1/genetics
5.
Cell Mol Life Sci ; 78(10): 4703-4712, 2021 May.
Article in English | MEDLINE | ID: mdl-33847763

ABSTRACT

Genetic analyses have revealed the pivotal contribution of microglial dysfunctions to the pathogenesis of Alzheimer's disease (AD). Along AD progression, the accumulation of danger-associated molecular patterns (DAMPs) including beta-amyloid and hyperphosphorylated tau continuously stimulates microglia, which results in their chronic activation. Chronically activated microglia secrete excessive pro-inflammatory cytokines, which further regulate microglial responses towards DAMPs. This has spurred longstanding interest in targeting cytokine-induced microglial responses for AD therapeutic development. However, the cytokine-induced microglial state transition is not comprehensively understood. Cytokines are assumed to induce microglial state transition from a resting state to an activated state. However, recent evidence indicate that this microglial state transition involves multiple sequential functional states. Moreover, the mechanisms by which different functional states within the cytokine-induced microglial state transition regulate AD pathology remain unclear. In this review, we summarize how different cytokine signaling pathways, including those of IL-33 (interleukin-33), NLRP3 inflammasome-IL-1ß, IL-10, and IL-12/IL-23, regulate microglial functions in AD. Furthermore, we discuss how the modulation of these cytokine signaling pathways can result in beneficial outcomes in AD. Finally, we describe a stepwise functional state transition of microglia induced by cytokine signaling that can provide insights into the molecular basis of the beneficial effects of cytokine modulation in AD and potentially aid therapeutic development.


Subject(s)
Alzheimer Disease/metabolism , Cytokines/metabolism , Microglia/metabolism , Signal Transduction/physiology , Amyloid beta-Peptides/metabolism , Animals , Humans , Inflammasomes/metabolism
6.
Sci Rep ; 11(1): 4359, 2021 02 23.
Article in English | MEDLINE | ID: mdl-33623128

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder with no disease-modifying treatment. AD progression is characterized by cognitive decline, neuroinflammation, and accumulation of amyloid-beta (Aß) and neurofibrillary tangles in the brain, leading to neuronal and glial dysfunctions. Neuropeptides govern diverse pathophysiological processes and represent key players in AD pathogenesis, regulating synaptic plasticity, glial cell functions and amyloid pathology. Activation of the pro-opiomelanocortin (POMC)-derived neuropeptide and its receptor from the melanocortin receptor (MCR) family have previously been shown to rescue the impairment in hippocampus-dependent synaptic plasticity in the APP/PS1 mouse model of AD. However, the functional roles of MCR signaling in AD conditions, particularly in glial functions, are largely unknown. In this study, we investigated the potential benefits of MCR activation in AD. In APP/PS1 transgenic mice, we demonstrate that MCR activation mediated by the central administration of its agonist D-Tyr MTII substantially reduces Aß accumulation, while alleviating global inflammation and astrocytic activation, particularly in the hippocampus. MCR activation prominently reduces the A1 subtype of reactive astrocytes, which is considered a key source of astrocytic neurotoxicity in AD. Concordantly, MCR activation suppresses microglial activation, while enhancing their association with amyloid plaques. The blunted activation of microglia may contribute to the reduction in the neurotoxic phenotypes of astrocytes. Importantly, transcriptome analysis reveals that MCR activation restores the impaired homeostatic processes and microglial reactivity in the hippocampus in APP/PS1 mice. Collectively, our findings demonstrate the potential of MCR signaling as therapeutic target for AD.


Subject(s)
Alzheimer Disease/drug therapy , Astrocytes/metabolism , Receptors, Melanocortin/agonists , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Inbred C57BL , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Peptides, Cyclic/chemistry , Receptors, Melanocortin/metabolism , Tyrosine/analogs & derivatives , alpha-MSH/analogs & derivatives , alpha-MSH/chemistry
7.
STAR Protoc ; 2(1): 100265, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33490981

ABSTRACT

Alzheimer's disease is characterized by the deposition of extracellular amyloid-beta (Aß) plaques. While microglial phagocytosis is a major mechanism through which Aß is cleared, there is no method for quantitatively assessing Aß phagocytic capacity of microglia in vivo. Here, we present a flow cytometry-based method for investigating the Aß phagocytic capacity of microglia in vivo. This method enables the direct comparison of Aß phagocytic capacity between different microglial subpopulations as well as the direct isolation of Aß phagocytic microglia for downstream applications. For complete details on the use and execution of this protocol, please refer to Lau et al. (2020).


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Microglia/immunology , Phagocytosis , Plaque, Amyloid/immunology , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , Plaque, Amyloid/genetics
8.
Proc Natl Acad Sci U S A ; 117(41): 25800-25809, 2020 10 13.
Article in English | MEDLINE | ID: mdl-32989152

ABSTRACT

Alzheimer's disease (AD) is the most common form of dementia but has no effective treatment. A comprehensive investigation of cell type-specific responses and cellular heterogeneity in AD is required to provide precise molecular and cellular targets for therapeutic development. Accordingly, we perform single-nucleus transcriptome analysis of 169,496 nuclei from the prefrontal cortical samples of AD patients and normal control (NC) subjects. Differential analysis shows that the cell type-specific transcriptomic changes in AD are associated with the disruption of biological processes including angiogenesis, immune activation, synaptic signaling, and myelination. Subcluster analysis reveals that compared to NC brains, AD brains contain fewer neuroprotective astrocytes and oligodendrocytes. Importantly, our findings show that a subpopulation of angiogenic endothelial cells is induced in the brain in patients with AD. These angiogenic endothelial cells exhibit increased expression of angiogenic growth factors and their receptors (i.e., EGFL7, FLT1, and VWF) and antigen-presentation machinery (i.e., B2M and HLA-E). This suggests that these endothelial cells contribute to angiogenesis and immune response in AD pathogenesis. Thus, our comprehensive molecular profiling of brain samples from patients with AD reveals previously unknown molecular changes as well as cellular targets that potentially underlie the functional dysregulation of endothelial cells, astrocytes, and oligodendrocytes in AD, providing important insights for therapeutic development.


Subject(s)
Alzheimer Disease/genetics , Cell Nucleus/genetics , Endothelial Cells/metabolism , Neuroglia/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Endothelial Cells/cytology , Female , Gene Expression Profiling , Humans , Male , Neuroglia/cytology , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Single-Cell Analysis , Transcriptome
9.
Cell Rep ; 31(3): 107530, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320664

ABSTRACT

Impairment of microglial clearance activity contributes to beta-amyloid (Aß) pathology in Alzheimer's disease (AD). While the transcriptome profile of microglia directs microglial functions, how the microglial transcriptome can be regulated to alleviate AD pathology is largely unknown. Here, we show that injection of interleukin (IL)-33 in an AD transgenic mouse model ameliorates Aß pathology by reprogramming microglial epigenetic and transcriptomic profiles to induce a microglial subpopulation with enhanced phagocytic activity. These IL-33-responsive microglia (IL-33RMs) express a distinct transcriptome signature that is highlighted by increased major histocompatibility complex class II genes and restored homeostatic signature genes. IL-33-induced remodeling of chromatin accessibility and PU.1 transcription factor binding at the signature genes of IL-33RM control their transcriptome reprogramming. Specifically, disrupting PU.1-DNA interaction abolishes the microglial state transition and Aß clearance that is induced by IL-33. Thus, we define a PU.1-dependent transcriptional pathway that drives the IL-33-induced functional state transition of microglia, resulting in enhanced Aß clearance.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Interleukin-33/pharmacology , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Disease Models, Animal , Female , Humans , Interleukin-33/genetics , Male , Mice , Mice, Transgenic , Microglia/pathology , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/pharmacology , Trans-Activators/metabolism , Transcriptome/drug effects
10.
Proc Natl Acad Sci U S A ; 116(27): 13651-13660, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209021

ABSTRACT

Adult hippocampal neurogenesis involves the lifelong generation of neurons. The process depends on the homeostasis of the production of neurons and maintenance of the adult neural stem cell (NSC) pool. Here, we report that α2-chimaerin, a Rho GTPase-activating protein, is essential for NSC homeostasis in adult hippocampal neurogenesis. Conditional deletion of α2-chimaerin in adult NSCs resulted in the premature differentiation of NSCs into intermediate progenitor cells (IPCs), which ultimately depleted the NSC pool and impaired neuron generation. Single-cell RNA sequencing and pseudotime analyses revealed that α2-chimaerin-conditional knockout (α2-CKO) mice lacked a unique NSC subpopulation, termed Klotho-expressing NSCs, during the transition of NSCs to IPCs. Furthermore, α2-CKO led to defects in hippocampal synaptic plasticity and anxiety/depression-like behaviors in mice. Our findings collectively demonstrate that α2-chimaerin plays an essential role in adult hippocampal NSC homeostasis to maintain proper brain function.


Subject(s)
Chimerin Proteins/physiology , GTP Phosphohydrolase Activators/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Animals , Cell Differentiation , Gene Knockdown Techniques , Hippocampus/physiology , Homeostasis , Mice , Mice, Knockout , Neural Stem Cells/physiology , Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...