Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yuka Atagi is active.

Publication


Featured researches published by Yuka Atagi.


Journal of Biological Chemistry | 2015

Apolipoprotein E is a Ligand for Triggering Receptor Expressed on Myeloid Cells 2 (TREM2)

Yuka Atagi; Chia Chen Liu; Meghan M. Painter; Xiao Fen Chen; Christophe Verbeeck; Honghua Zheng; Xia Li; Rosa Rademakers; Silvia S. Kang; Huaxi Xu; Steven G. Younkin; Pritam Das; John D. Fryer; Guojun Bu

Background: TREM2 is associated with several neurodegenerative diseases. Results: ApoE bound to TREM2 and increased phagocytosis of apoptotic neurons by microglia. Alzheimer disease (AD) risk-associated TREM2-R47H mutant had a reduced binding to apoE. Conclusion: ApoE is a novel ligand for TREM2. Interaction between apoE and TREM2 likely regulates phagocytosis of apoE-bound apoptotic neurons. Significance: Interaction between two AD risk-associated proteins modulates microglial function. Several heterozygous missense mutations in the triggering receptor expressed on myeloid cells 2 (TREM2) have recently been linked to risk for a number of neurological disorders including Alzheimer disease (AD), Parkinson disease, and frontotemporal dementia. These discoveries have re-ignited interest in the role of neuroinflammation in the pathogenesis of neurodegenerative diseases. TREM2 is highly expressed in microglia, the resident immune cells of the central nervous system. Along with its adaptor protein, DAP12, TREM2 regulates inflammatory cytokine release and phagocytosis of apoptotic neurons. Here, we report apolipoprotein E (apoE) as a novel ligand for TREM2. Using a biochemical assay, we demonstrated high-affinity binding of apoE to human TREM2. The functional significance of this binding was highlighted by increased phagocytosis of apoE-bound apoptotic N2a cells by primary microglia in a manner that depends on TREM2 expression. Moreover, when the AD-associated TREM2-R47H mutant was used in biochemical assays, apoE binding was vastly reduced. Our data demonstrate that apoE-TREM2 interaction in microglia plays critical roles in modulating phagocytosis of apoE-bound apoptotic neurons and establish a critical link between two proteins whose genes are strongly linked to the risk for AD.


Journal of Experimental Medicine | 2017

Soluble TREM2 induces inflammatory responses and enhances microglial survival

Li Zhong; Xiao Fen Chen; Tingting Wang; Zhe Wang; Chunyan Liao; Zongqi Wang; Ruizhi Huang; Daxin Wang; Xinxiu Li; Linbei Wu; Lin Jia; Honghua Zheng; Meghan M. Painter; Yuka Atagi; Chia Chen Liu; Yun Wu Zhang; John D. Fryer; Huaxi Xu; Guojun Bu

Triggering receptor expressed on myeloid cells 2 (TREM2) is an innate immune receptor expressed in microglia in the brain. A soluble form of TREM2 (sTREM2) derived from proteolytic cleavage of the cell surface receptor is increased in the preclinical stages of AD and positively correlates with the amounts of total and phosphorylated tau in the cerebrospinal fluid. However, the physiological and pathological functions of sTREM2 remain unknown. Here, we show that sTREM2 promotes microglial survival in a PI3K/Akt-dependent manner and stimulates the production of inflammatory cytokines depending on NF-&kgr;B. Variants of sTREM2 carrying AD risk-associated mutations were less potent in both suppressing apoptosis and triggering inflammatory responses. Importantly, sTREM2 delivered to the hippocampi of both wild-type and Trem2-knockout mice elevated the expression of inflammatory cytokines and induced morphological changes of microglia. Collectively, these data indicate that sTREM2 triggers microglial activation inducing inflammatory responses and promoting survival. This study has implications for the pathogenesis of AD and provides insights into targeting sTREM2 pathway for AD therapy.


Journal of Biological Chemistry | 2015

Apolipoprotein E Inhibits Cerebrovascular Pericyte Mobility through a RhoA Protein-mediated Pathway

Caroline S. Casey; Yuka Atagi; Yu Yamazaki; Mitsuru Shinohara; Masaya Tachibana; Yuan Fu; Guojun Bu; Takahisa Kanekiyo

Background: Pericytes are a major cerebrovasculature component, producing abundant apolipoprotein E (apoE). Results: Deletion of apoE accelerates pericyte migration and adhesion by increasing RhoA activity. The effects are reversed by exogenous apoE3 but not apoE4. Conclusion: ApoE suppresses pericyte mobility in a RhoA-mediated manner. Significance: Pericyte-derived apoE regulates cell mobility and can be explored as a target for neurovascular disease therapy. Pericytes play a critical role in the cerebrovasculature within the CNS. These small contractile cells produce large quantities of apolipoprotein E (apoE) whose isoforms influence cerebrovascular functions and determine the genetic risk for Alzheimer disease. Despite extensive studies on astrocyte-secreted apoE, which supports synapses by transporting cholesterol to neurons, the biochemical properties and function of apoE secreted by pericytes are not clear. Because pericytes mediate important functions in the CNS, including the initiation of glial scar formation, angiogenesis, and maintenance of the blood-brain barrier, we investigated the potential role of apoE in pericyte mobility. We found that knockdown of apoE expression significantly accelerates pericyte migration, an effect that can be rescued by exogenous apoE3, but not apoE4, a risk factor for Alzheimer disease. ApoE-regulated migration of pericytes also requires the function of the low-density lipoprotein receptor-related protein 1 (LRP1), a major apoE receptor in the brain that is abundantly expressed in pericytes. Because apoE-knockdown also leads to enhanced cell adhesion, we investigated the role of apoE in the regulation of the actin cytoskeleton. Interestingly, we found that the levels of active RhoA are increased significantly in apoE knockdown pericytes and that RhoA inhibitors blocked pericyte migration. Taken together, our results suggest that apoE has an intrinsic role in pericyte mobility, which is vital in maintaining cerebrovascular function. These findings provide novel insights into the role of apoE in the cerebrovascular system.


Molecular Neurodegeneration | 2016

Apolipoprotein E lipoprotein particles inhibit amyloid-β uptake through cell surface heparan sulphate proteoglycan

Yuan Fu; Jing Zhao; Yuka Atagi; Henrietta M. Nielsen; Chia Chen Liu; Honghua Zheng; Mitsuru Shinohara; Takahisa Kanekiyo; Guojun Bu

BackgroundThe accumulation, aggregation and deposition of amyloid-β (Aβ) peptides in the brain are central to the pathogenesis of Alzheimer’s disease (AD). Alzheimer’s disease risk increases significantly in individuals carrying one or two copies of APOE ε4 allele compared to individuals with an ε3/ε3 genotype. Growing evidence has demonstrated that apolipoprotein E (apoE) strongly influences AD pathogenesis by controlling Aβ aggregation and metabolism. Heparan sulphate proteoglycans (HSPGs) are abundant cell surface molecules that bind to both apoE and Aβ. HSPGs have been associated with Aβ aggregation and deposition. Although several lines of research have shown that apoE influences Aβ clearance in the brain, it is not clear how apoE influences HSPG-mediated cellular uptake of Aβ.ResultsIn this study, we show that apoE lipoprotein particles from conditioned media of immortalized astrocytes isolated from human APOE-targeted replacement (TR) mice significantly suppress cellular Aβ42 and Aβ40 uptake through cell surface HSPG. ApoE3 and apoE4 particles have similar binding affinity to heparin, while apoE4 particles are likely hypolipidated compared to apoE particles. We also found that the apoE particles antagonize Aβ binding to cell surface, and inhibited Aβ uptake in a concentration-dependent manner in Chinese hamster ovary (CHO) cells. While the effect was not apoE isoform-dependent, the suppressive effect of apoE particles on Aβ uptake was not observed in HSPG-deficient CHO cells. We further demonstrated that apoE particles reduced the internalization of Aβ in mouse primary neurons, an effect that is eliminated by the presence of heparin.ConclusionsTaken together, our findings indicate that apoE particles irrespective of isoform inhibit HSPG-dependent cellular Aβ uptake. Modulating the ability of apoE particles to affect Aβ cellular uptake may hold promises for developing new strategies for AD therapy.


Journal of Neuroinflammation | 2016

LRP1 modulates the microglial immune response via regulation of JNK and NF-κB signaling pathways

Longyu Yang; Chia-Chen Liu; Honghua Zheng; Takahisa Kanekiyo; Yuka Atagi; Lin Jia; Daxin Wang; Aurelie N’Songo; Dan Can; Huaxi Xu; Xiao-Fen Chen; Guojun Bu

BackgroundNeuroinflammation is characterized by microglial activation and the increased levels of cytokines and chemokines in the central nervous system (CNS). Recent evidence has implicated both beneficial and toxic roles of microglia when over-activated upon nerve injury or in neurodegenerative diseases, including Alzheimer’s disease (AD). The low-density lipoprotein receptor-related protein 1 (LRP1) is a major receptor for apolipoprotein E (apoE) and amyloid-β (Aβ), which play critical roles in AD pathogenesis. LRP1 regulates inflammatory responses in peripheral tissues by modulating the release of inflammatory cytokines and phagocytosis. However, the roles of LRP1 in brain innate immunity and neuroinflammation remain unclear.MethodsIn this study, we determined whether LRP1 modulates microglial activation by knocking down Lrp1 in mouse primary microglia. LRP1-related functions in microglia were also assessed in the presence of LRP1 antagonist, the receptor-associated protein (RAP). The effects on the production of inflammatory cytokines were measured by quantitative real-time PCR (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA). Potential involvement of specific signaling pathways in LRP1-regulated functions including mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB) were assessed using specific inhibitors.ResultsWe found that knocking down of Lrp1 in mouse primary microglia led to the activation of both c-Jun N-terminal kinase (JNK) and NF-κB pathways with corresponding enhanced sensitivity to lipopolysaccharide (LPS) in the production of pro-inflammatory cytokines. Similar effects were observed when microglia were treated with LRP1 antagonist RAP. In addition, treatment with pro-inflammatory stimuli suppressed Lrp1 expression in microglia. Interestingly, NF-κB inhibitor not only suppressed the production of cytokines induced by the knockdown of Lrp1 but also restored the down-regulated expression of Lrp1 by LPS.ConclusionsOur study uncovers that LRP1 suppresses microglial activation by modulating JNK and NF-κB signaling pathways. Given that dysregulation of LRP1 has been associated with AD pathogenesis, our work reveals a critical regulatory mechanism of microglial activation by LRP1 that could be associated with other AD-related pathways thus further nominating LRP1 as a potential disease-modifying target for the treatment of AD.


Molecular Neurodegeneration | 2015

TREM2 in CNS homeostasis and neurodegenerative disease

Meghan M. Painter; Yuka Atagi; Chia Chen Liu; Rosa Rademakers; Huaxi Xu; John D. Fryer; Guojun Bu


Acta Neuropathologica | 2014

Total apolipoprotein E levels and specific isoform composition in cerebrospinal fluid and plasma from Alzheimer's disease patients and controls

Eduardo Martínez-Morillo; Oskar Hansson; Yuka Atagi; Guojun Bu; Lennart Minthon; Eleftherios P. Diamandis; Henrietta M. Nielsen


Neurobiology of Aging | 2016

Opposing roles of the triggering receptor expressed on myeloid cells 2 and triggering receptor expressed on myeloid cells-like transcript 2 in microglia activation

Honghua Zheng; Chia Chen Liu; Yuka Atagi; Xiao Fen Chen; Lin Jia; Longyu Yang; Wencan He; Xilin Zhang; Silvia S. Kang; Terrone L. Rosenberry; John D. Fryer; Yun Wu Zhang; Huaxi Xu; Guojun Bu


Acta Neuropathologica | 2016

Impact of sex and APOE4 on cerebral amyloid angiopathy in Alzheimer’s disease

Mitsuru Shinohara; Melissa E. Murray; Ryan D. Frank; Motoko Shinohara; Michael DeTure; Yu Yamazaki; Masaya Tachibana; Yuka Atagi; Mary D. Davis; Chia Chen Liu; Na Zhao; Meghan M. Painter; Ronald C. Petersen; John D. Fryer; Julia E. Crook; Dennis W. Dickson; Guojun Bu; Takahisa Kanekiyo


Molecular Neurodegeneration | 2015

Frontotemporal dementia-associated N279K tau mutant disrupts subcellular vesicle trafficking and induces cellular stress in iPSC-derived neural stem cells

Melissa C. Wren; Jing Zhao; Chia Chen Liu; Melissa E. Murray; Yuka Atagi; Mary D. Davis; Yuan Fu; Hirotaka James Okano; Kotaro Ogaki; Audrey Strongosky; Pawel Tacik; Rosa Rademakers; Owen A. Ross; Dennis W. Dickson; Zbigniew K. Wszolek; Takahisa Kanekiyo; Guojun Bu

Collaboration


Dive into the Yuka Atagi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge