Youngdae Gwon
Seoul National University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Youngdae Gwon.
Journal of Clinical Investigation | 2013
Tae In Kam; Sungmin Song; Youngdae Gwon; Hyejin Park; Ji Jing Yan; Isak Im; Ji Woo Choi; Tae-Yong Choi; Jeongyeon Kim; Dong Keun Song; Toshiyuki Takai; Yong Chul Kim; Key Sun Kim; Se-Young Choi; Sukwoo Choi; William L. Klein; Junying Yuan; Yong-Keun Jung
Amyloid-β (Aβ) induces neuronal loss and cognitive deficits and is believed to be a prominent cause of Alzheimers disease (AD); however, the cellular pathology of the disease is not fully understood. Here, we report that IgG Fcγ receptor II-b (FcγRIIb) mediates Aβ neurotoxicity and neurodegeneration. We found that FcγRIIb is significantly upregulated in the hippocampus of AD brains and neuronal cells exposed to synthetic Aβ. Neuronal FcγRIIb activated ER stress and caspase-12, and Fcgr2b KO primary neurons were resistant to synthetic Aβ-induced cell death in vitro. Fcgr2b deficiency ameliorated Aβ-induced inhibition of long-term potentiation and inhibited the reduction of synaptic density by naturally secreted Aβ. Moreover, genetic depletion of Fcgr2b rescued memory impairments in an AD mouse model. To determine the mechanism of action of FcγRIIb in Aβ neurotoxicity, we demonstrated that soluble Aβ oligomers interact with FcγRIIb in vitro and in AD brains, and that inhibition of their interaction blocks synthetic Aβ neurotoxicity. We conclude that FcγRIIb has an aberrant, but essential, role in Aβ-mediated neuronal dysfunction.
Human Molecular Genetics | 2012
Hyejin Park; Tae-In Kam; Youngdoo Kim; Hyunwoo Choi; Youngdae Gwon; Changsoo Kim; Jae-Young Koh; Yong-Keun Jung
Abnormally hyperphosphorylated tau is often caused by tau kinases, such as GSK3β and Cdk5. Such occurrence leads to neurofibrillary tangle formation and neuronal degeneration in tauopathy, including Alzheimers disease (AD). However, little is known about the signaling cascade underlying the pathologic phosphorylation of tau by Aβ(42). In this study, we show that adenylate kinase 1 (AK1) is a novel regulator of abnormal tau phosphorylation. AK1 expression is markedly increased in the brains of AD patients and AD model mice and is significantly induced by Aβ(42) in the primary neurons. Ectopic expression of AK1 alone augments the pathologic phosphorylation of tau at PHF1, CP13 and AT180 epitopes and enhances the formation of tau aggregates. Inversely, downregulation of AK1 alleviates Aβ(42)-induced hyperphosphorylation of tau. AK1 plays a role in Aβ(42)-induced impairment of AMPK activity and GSK3β activation in the primary neurons. Pharmacologic studies show that treatment with an AMPK inhibitor activates GSK3β, and a GSK3β inhibitor attenuates AK1-mediated tau phosphorylation. In a Drosophila model of human tauopathy, the retinal expression of human AK1 severely exacerbates rough eye phenotype and increases abnormal tau phosphorylation. Further, neural expression of AK1 reduces the lifespan of tau transgenic files. Taken together, these observations indicate that the neuronal expression of AK1 is induced by Aβ(42) to increase abnormal tau phosphorylation via AMPK-GSK3β and contributes to tau-mediated neurodegeneration, providing a new upstream modulator of GSK3β in the pathologic phosphorylation of tau.
Cellular and Molecular Life Sciences | 2014
Tae-In Kam; Youngdae Gwon; Yong-Keun Jung
Alzheimer’s disease (AD) is the most common neurodegenerative disease. Although a major cause of AD is the accumulation of amyloid-β (Aβ) peptide that induces neuronal loss and cognitive impairments, our understanding of its neurotoxic mechanisms is limited. Recent studies have identified putative Aβ-binding receptors that mediate Aβ neurotoxicity in cells and models of AD. Once Aβ interacts with a receptor, a toxic signal is transduced into neurons, resulting in cellular defects including endoplasmic reticulum stress and mitochondrial dysfunction. In addition, Aβ can also be internalized into neurons through unidentified Aβ receptors and induces malfunction of subcellular organelles, which explains some part of Aβ neurotoxicity. Understanding the neurotoxic signaling initiated by Aβ-receptor binding and cellular defects provide insight into new therapeutic windows for AD. In the present review, we summarize the findings on Aβ-binding receptors and the neurotoxicity of oligomeric Aβ.
eLife | 2016
Tae-In Kam; Hyejin Park; Youngdae Gwon; Sungmin Song; Seo-Hyun Kim; Seo Won Moon; Dong-Gyu Jo; Yong-Keun Jung
Amyloid-β (Aβ)-containing extracellular plaques and hyperphosphorylated tau-loaded intracellular neurofibrillary tangles are neuropathological hallmarks of Alzheimers disease (AD). Although Aβ exerts neuropathogenic activity through tau, the mechanistic link between Aβ and tau pathology remains unknown. Here, we showed that the FcγRIIb-SHIP2 axis is critical in Aβ1-42-induced tau pathology. Fcgr2b knockout or antagonistic FcγRIIb antibody inhibited Aβ1-42-induced tau hyperphosphorylation and rescued memory impairments in AD mouse models. FcγRIIb phosphorylation at Tyr273 was found in AD brains, in neuronal cells exposed to Aβ1-42, and recruited SHIP2 to form a protein complex. Consequently, treatment with Aβ1-42 increased PtdIns(3,4)P2 levels from PtdIns(3,4,5)P3 to mediate tau hyperphosphorylation. Further, we found that targeting SHIP2 expression by lentiviral siRNA in 3xTg-AD mice or pharmacological inhibition of SHIP2 potently rescued tau hyperphosphorylation and memory impairments. Thus, we concluded that the FcγRIIb-SHIP2 axis links Aβ neurotoxicity to tau pathology by dysregulating PtdIns(3,4)P2 metabolism, providing insight into therapeutic potential against AD. DOI: http://dx.doi.org/10.7554/eLife.18691.001
The Journal of Neuroscience | 2018
Youngdae Gwon; Tae-In Kam; Seo-Hyun Kim; Sungmin Song; Hyejin Park; Bitna Lim; Haneul Lee; Weontae Lee; Dong-Gyu Jo; Yong-Keun Jung
Emerging evidences suggest that intraneuronal Aβ correlates with the onset of Alzheimers disease (AD) and highly contributes to neurodegeneration. However, critical mediator responsible for Aβ uptake in AD pathology needs to be clarified. Here, we report that FcγRIIb2, a variant of Fcγ-receptor IIb (FcγRIIb), functions in neuronal uptake of pathogenic Aβ. Cellular accumulation of oligomeric Aβ1–42, not monomeric Aβ1–42 or oligomeric Aβ1–40, was blocked by Fcgr2b knock-out in neurons and partially in astrocytes. Aβ1–42 internalization was FcγRIIb2 di-leucine motif-dependent and attenuated by TOM1, a FcγRIIb2-binding protein that repressed the receptor recycling. TOM1 expression was downregulated in the hippocampus of male 3xTg-AD mice and AD patients, and regulated by miR-126-3p in neuronal cells after exposure to Aβ1–42. In addition, memory impairments in male 3xTg-AD mice were rescued by the lentiviral administration of TOM1 gene. Augmented Aβ uptake into lysosome caused its accumulation in cytoplasm and mitochondria. Moreover, neuronal accumulation of Aβ in both sexes of 3xTg-AD mice and memory deficits in male 3xTg-AD mice were ameliorated by forebrain-specific expression of Aβ-uptake-defective Fcgr2b mutant. Our findings suggest that FcγRIIb2 is essential for neuropathic uptake of Aβ in AD. SIGNIFICANCE STATEMENT Accumulating evidences suggest that intraneuronal Aβ is found in the early step of AD brain and is implicated in the pathogenesis of AD. However, the critical mediator involved in these processes is uncertain. Here, we describe that the FcγRIIb2 variant is responsible for both neuronal uptake and intraneuronal distribution of pathogenic Aβ linked to memory deficits in AD mice, showing a pathologic significance of the internalized Aβ. Further, Aβ internalization is attenuated by TOM1, a novel FcγRIIb2-binding protein. Together, we provide a molecular mechanism responsible for neuronal uptake of pathogenic Aβ found in AD.
Alzheimers & Dementia | 2017
Seo-Hyun Kim; Tae-In Kam; Youngdae Gwon; Yong-Keun Jung
(AD-DS). Three copies of the Hsa21 gene APP are sufficient to cause early-onset AD but how trisomy of other Hsa21 genes influences disease development is unclear. Methods:We analysed cathepsin activity in patient fibroblasts, post-mortem brain material from people who have DS and AD pathology and a novel mouse model of AD-DS (progeny of the cross of the Tc1 model of Hsa21 trisomy with the J20 APP transgenic model). To understand how changes in cathepsin activity may affect APP and Ab processing in vitro and in vivo in our AD-DS animal model, we used a combination of pulse-chase analysis, western blotting, immunohistochemistry, ELISA, mass-spectrometry, enzymatic activity assays and in vivo Ab clearance studies. Additionally we undertook behavioural phenotyping and an aging study of our mouse model of AD-DS to understand how changes in APP/Ab processing due to trisomy of Hsa21 effect learning and aging. Results:Here we show that triplication of Hsa21 sequences, other than APP, cause cysteine cathepsin deficits that result in failure to activate these proteases in DS. We successfully modelled these enzymatic changes in a novel AD-DS mouse model system, and found that they occur independently of gross-enlargement of the endo-lysosomes. Using our AD-DS mouse model, we show that trisomy of Hsa21 sequences, other than APP, also alter the metabolism of APP/Ab. These changes decrease the soluble Ab38/42 ratio and are associated with an increase in Ab aggregation and deposition, and result in exacerbation of APP/Ab-associated hyper-activity and specific deficits in two tests of short-term memory. We also show that the trisomyassociated changes in APP/Ab metabolism we observe occur independently of alterations in a-, bor g-secretase activity or changes in the rate of extracellular Ab-clearance in vivo. Conclusions:We propose that trisomy Hsa21-associated cathepsin deficits are a novel AD-DS pathomechanism that alter APP/Ab processing and may contribute to the development of AD in people who have DS.
Alzheimers & Dementia | 2010
Tae-In Kam; Sungmin Song; Youngdae Gwon; Yong-Keun Jung
Background: Alzheimer’s disease (AD) is associated with synaptic loss and memory impairment. AD transgenic mice (Tg) models, over-expressing mutated forms of the amyloid precursor protein demonstrate memory deficit and inhibition of the long-term potentiation prior to the appearance of Aß plaques, which implicates toxicity of soluble Aß oligomeric species in the early pathogenesis of memory dysfunction. Experiments in primary cultures of hippocampal neurons showed preferential binding of Aß oligomers to Nmethyl-D-aspartic acid (NMDA) receptors resulting over time in their down regulation. NMDA receptor signaling involves Ca for intracellular signal transduction and is pivotal for the synaptic plasticity in the hippocampus, which underlies memory formation. The goal of our experiments was to characterize impairment of NMDA receptor signaling AD Tg mice, which has not been done before. Methods: NMDA receptor signaling in the CA1 sector of the hippocampus was investigated using microdialysis in awake and behaving 3.5-4 month old APPSW/PS1dE9 AD Tg mice. Under physiological conditions stimulation of NMDA receptor results in a massive increase in the intracellular Ca concentration, which via Ca/ calmodulin / nitric oxide synthase / nitric oxide / soluble guanyl cyclase pathway stimulates production of cGMP. Thus cGMP released to the interstitial fluid can serve as a measure of NMDA receptor function. Results: Stimulation of the NMDA receptor with 500mM NMDA (a selective NMDA agonist) administered through reverse microdialysis resulted in 32% increase in the baseline cGMP level in four month old wild type mice (p < 0.05, paired ttest). In contrast, only 2%, non-significant change in 3.5-4 month old APPSW/PS1dE9 Tg mice was noticed (n 1⁄4 8). Examination of APPSW/ PS1dE9 Tg mice brains revealed minimal Aß plaque load in the CA1 hippocampal sector. These mice had high tissue concentration of Aß species, with predominance of oligomerization prone Aß42. Conclusions: Our experiments provide direct proof of the NMDA receptor signaling impairment in AD Tg mice, which is associated with presence of Aß oligomers but not Aß deposits. Biochemical analysis of NMDA receptor and other post-synaptic proteins is close to explaining whether impairment of the NMDA receptor signaling is associated with down regulation of its expression.
Alzheimers & Dementia | 2018
Youngdae Gwon; Tae-In Kam; Seo-Hyun Kim; Sungmin Song; Hyejin Park; Bitna Lim; Ha-Neul Lee; Weontae Lee; Dong-Gyu Jo; Yong-Keun Jung
한국실험동물학회 학술발표대회 논문집 | 2015
Tae-In Kam; Sungmin Song; Youngdae Gwon; Hee Sae Park; Yong-Keun Jung
Archive | 2012
Hyejin Park; Tae-In Kam; Youngdoo Kim; Hyunwoo Choi; Youngdae Gwon; Changsoo Kim; Jae-Young Koh; Yong-Keun Jung