Network


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

Hotspot


Dive into the research topics where Sang-Won Min is active.

Publication


Featured researches published by Sang-Won Min.


Nature Medicine | 2015

Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits

Sang-Won Min; Xu Chen; Tara E. Tracy; Yaqiao Li; Yungui Zhou; Chao Wang; Kotaro Shirakawa; S. Sakura Minami; Erwin Defensor; Sue-Ann Mok; Peter Dongmin Sohn; Birgit Schilling; Xin Cong; Bradford W. Gibson; Jeffrey R. Johnson; Nevan J. Krogan; Mehrdad Shamloo; Jason E. Gestwicki; Eliezer Masliah; Eric Verdin; Li Gan

Tauopathies, including frontotemporal dementia (FTD) and Alzheimers 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.


Proceedings of the National Academy of Sciences of the United States of America | 2007

E-Syts, a family of membranous Ca2+-sensor proteins with multiple C2 domains

Sang-Won Min; Wen-Pin Chang; Thomas C. Südhof

C2 domains are autonomously folded protein modules that generally act as Ca2+- and phospholipid-binding domains and/or as protein–protein interaction domains. We now report the primary structures and biochemical properties of a family of evolutionarily conserved mammalian proteins, referred to as E-Syts, for extended synaptotagmin-like proteins. E-Syts contain an N-terminal transmembrane region, a central juxtamembranous domain that is conserved from yeast to human, and five (E-Syt1) or three (E-Syt2 and E-Syt3) C-terminal C2 domains. Only the first E-Syt C2 domain, the C2A domain, includes the complete sequence motif that is required for Ca2+ binding in C2 domains. Recombinant protein fragments of E-Syt2 that include the first C2 domain are capable of Ca2+-dependent phospholipid binding at micromolar concentrations of free Ca2+, suggesting that E-Syts bind Ca2+ through their first C2 domain in a phospholipid complex. E-Syts are ubiquitously expressed, but enriched in brain. Expression of myc-tagged E-Syt proteins in transfected cells demonstrated localization to intracellular membranes for E-Syt1 and to plasma membranes for E-Syt2 and E-Syt3. Structure/function studies showed that the plasma-membrane localization of E-Syt2 and E-Syt3 was directed by their C-terminal C2C domains. This result reveals an unexpected mechanism by which the C2C domains of E-Syt2 and E-Syt3 functions as a targeting motif that localizes these proteins into the plasma membrane independent of their transmembrane region. Viewed together, our findings suggest that E-Syts function as Ca2+-regulated intrinsic membrane proteins with multiple C2 domains, expanding the repertoire of such proteins to a fourth class beyond synaptotagmins, ferlins, and MCTPs (multiple C2 domain and transmembrane region proteins).


Nature Medicine | 2014

Progranulin protects against amyloid β deposition and toxicity in Alzheimer's disease mouse models

S. Sakura Minami; Sang-Won Min; Grietje Krabbe; Chao Wang; Yungui Zhou; Rustam Asgarov; Yaqiao Li; Lauren Herl Martens; Lisa P Elia; Michael E. Ward; Lennart Mucke; Robert V. Farese; Li Gan

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 Alzheimers disease (AD). However, the role of PGRN in AD pathogenesis is unknown. Here we show that PGRN inhibits amyloid β (Aβ) 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. Aβ plaque load correlated negatively with levels of hippocampal PGRN, showing the dose-dependent inhibitory effects of PGRN on plaque deposition. PGRN also protected against Aβ toxicity. Lentivirus-mediated PGRN overexpression prevented spatial memory deficits and hippocampal neuronal loss in AD mice. The protective effects of PGRN against Aβ deposition and toxicity have important therapeutic implications. We propose enhancing PGRN as a potential treatment for PGRN-deficient FTLD and AD.


Frontiers in Aging Neuroscience | 2013

Sirtuins in neurodegenerative diseases: an update on potential mechanisms.

Sang-Won Min; Peter Dongmin Sohn; Seo-hyun Cho; Raymond A. Swanson; Li Gan

Silent information regulator 2 proteins (sirtuins or SIRTs) are a group of deacetylases (or deacylases) whose activities are dependent on and regulated by nicotinamide adenine dinucleotide (NAD+). Compelling evidence supports that sirtuins play major roles in many aspects of physiology, especially in pathways related to aging – the predominant and unifying risk factor for neurodegenerative diseases. In this review, we highlight the molecular mechanisms underlying the protective effects of sirtuins in neurodegenerative diseases, focusing on protein homeostasis, neural plasticity, mitochondrial function, and sustained chronic inflammation. We will also examine the potential and challenges of targeting sirtuin pathways to block these pathogenic pathways.


The Journal of Neuroscience | 2013

Titration of Syntaxin1 in Mammalian Synapses Reveals Multiple Roles in Vesicle Docking, Priming, and Release Probability

Marife Arancillo; Sang-Won Min; Stefan H. Gerber; Agnieszka Münster-Wandowski; Yuan-Ju Wu; Melissa A. Herman; Thorsten Trimbuch; Jong-Cheol Rah; Gudrun Ahnert-Hilger; Dietmar Riedel; Thomas C. Südhof; Christian Rosenmund

Synaptic vesicles undergo sequential steps in preparation for neurotransmitter release. Individual SNARE proteins and the SNARE complex itself have been implicated in these processes. However, discrete effects of SNARE proteins on synaptic function have been difficult to assess using complete loss-of-function approaches. We therefore used a genetic titration technique in cultured mouse hippocampal neurons to evaluate the contribution of the neuronal SNARE protein Syntaxin1 (Stx1) in vesicle docking, priming, and release probability. We generated graded reductions of total Stx1 levels by combining two approaches, namely, endogenous hypomorphic expression of the isoform Stx1B and RNAi-mediated knockdown. Proximity of synaptic vesicles to the active zone was not strongly affected. However, overall release efficiency of affected neurons was severely impaired, as demonstrated by a smaller readily releasable pool size, slower refilling rate of primed vesicles, and lower release probability. Interestingly, dose–response fitting of Stx1 levels against readily releasable pool size and vesicular release probability showed similar Kd (dissociation constant) values at 18% and 19% of wild-type Stx1, with cooperativity estimates of 3.4 and 2.5, respectively. This strongly suggests that priming and vesicle fusion share the same molecular stoichiometry, and are governed by highly related mechanisms.


Journal of Biological Chemistry | 2017

An inhibitor of the proteasomal deubiquitinating enzyme USP14 induces tau elimination in cultured neurons

Monica Boselli; Byung-Hoon Lee; Jessica Robert; Miguel A. Prado; Sang-Won Min; Chialin Cheng; M. Catarina Silva; Changhyun Seong; Suzanne Elsasser; Ketki M. Hatle; Timothy C. Gahman; Steven P. Gygi; Stephen J. Haggarty; Li Gan; Randall W. King; Daniel Finley

The ubiquitin-proteasome system (UPS) is responsible for most selective protein degradation in eukaryotes and regulates numerous cellular processes, including cell cycle control and protein quality control. A component of this system, the deubiquitinating enzyme USP14, associates with the proteasome where it can rescue substrates from degradation by removal of the ubiquitin tag. We previously found that a small-molecule inhibitor of USP14, known as IU1, can increase the rate of degradation of a subset of proteasome substrates. We report here the synthesis and characterization of 87 variants of IU1, which resulted in the identification of a 10-fold more potent USP14 inhibitor that retains specificity for USP14. The capacity of this compound, IU1-47, to enhance protein degradation in cells was tested using as a reporter the microtubule-associated protein tau, which has been implicated in many neurodegenerative diseases. Using primary neuronal cultures, IU1-47 was found to accelerate the rate of degradation of wild-type tau, the pathological tau mutants P301L and P301S, and the A152T tau variant. We also report that a specific residue in tau, lysine 174, is critical for the IU1-47–mediated tau degradation by the proteasome. Finally, we show that IU1-47 stimulates autophagic flux in primary neurons. In summary, these findings provide a powerful research tool for investigating the complex biology of USP14.


The Journal of Neuroscience | 2018

SIRT1 Deacetylates Tau and Reduces Pathogenic Tau Spread in a Mouse Model of Tauopathy

Sang-Won Min; Peter Dongmin Sohn; Yaqiao Li; Nino Devidze; Jeffrey R. Johnson; Nevan J. Krogan; Eliezer Masliah; Sue-Ann Mok; Jason E. Gestwicki; Li Gan

Hyperacetylation of tau has been implicated in neurodegeneration and cognitive decline in tauopathy brains. The nicotinamide adenosine dinucleotide-dependent class-III protein deacetylase SIRT1 is one of the major enzymes involved in removal of acetyl groups from tau in vitro. However, whether SIRT1 regulates acetylation of pathogenic tau and ameliorates tau-mediated pathogenesis remains unclear. Here, we report deacetylating activity of SIRT1 for acetylated Lys174 (K174) of tau in tauP301S transgenic mice with a brain-specific SIRT1 deletion. We show that SIRT1 deficiency leads to exacerbation of premature mortality, synapse loss, and behavioral disinhibition in tauP301S transgenic mice of both sexes. By contrast, SIRT1 overexpression by stereotaxic delivery of adeno-associated virus that encodes SIRT1 into the hippocampus reduces acetylated K174 tau. Furthermore, SIRT1 overexpression significantly attenuates the spread of tau pathology into anatomically connected brain regions of tauP301S transgenic mice of both sexes. These findings suggest the functional importance of SIRT1 in regulating pathogenic tau acetylation and in suppressing the spread of tau pathology in vivo. SIGNIFICANCE STATEMENT In neurodegenerative disorders with inclusions of microtubule-associated protein tau, aberrant lysine acetylation of tau plays critical roles in promoting tau accumulation and toxicity. Identifying strategies to deacetylate tau could interfere with disease progression; however, little is known about how pathogenic tau is deacetylated in vivo. Here we show that the protein deacetylase SIRT1 reduces tau acetylation in a mouse model of neurodegeneration. SIRT1 deficiency in the brain aggravates synapse loss and behavioral disinhibition, and SIRT1 overexpression ameliorates propagation of tau pathology.


Alzheimer's & Dementia: Translational Research & Clinical Interventions | 2017

An 8-week, open-label, dose-finding study of nimodipine for the treatment of progranulin insufficiency from GRN gene mutations

Sharon Sha; Zachary A. Miller; Sang-Won Min; Yungui Zhou; Jesse A. Brown; Laura L. Mitic; Anna Karydas; Mary Koestler; Richard Tsai; Chiara Corbetta-Rastelli; S. Lin; Emma Hare; Scott Fields; Kirsten E. Fleischmann; Ryan Powers; Ryan Fitch; Lauren Herl Martens; Mehrdad Shamloo; Anne M. Fagan; Robert V. Farese; Rodney Pearlman; William W. Seeley; Bruce L. Miller; Li Gan; Adam L. Boxer

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.


Alzheimers & Dementia | 2011

Acetylation of tau contributes to tau accumulation and dysfunction

Li Gan; Sang-Won Min; Seo-hyun Cho; Yungui Zhou; Vahram Haroutunian; Bill Seeley; Eric J. Huang; Yong Shen; Eliezer Masliah; Philip A. Cole; Melanie Ott

O4-04-01 ACETYLATION OF TAU CONTRIBUTES TO TAU ACCUMULATION AND DYSFUNCTION Li Gan, Sang-Won Min, Seo-hyun Cho, Yungui Zhou, Vahram Haroutunian, Bill Seeley, Eric Huang, Yong Shen, Eliezer Masliah, Philip Cole, Melanie Ott, Li Gan, Gladstone Institute of Neurological Disease, San Francisco, Calif, United States; Gladstone Insititute of Neurological Disease, UCSF, San Francisco, Calif, United States; 3 Gladstone Institute of Neurological Disease, UCSF, San Francisco, Calif, United States; 4 Mount Sinai School of Medicine, New York, New York, United States; UCSF, San Francisco, Calif, United States; Roskamp Institute, Sarasota, Florida, United States; UCSD, La Jolla, Calif, United States; 8 Johns Hopkins University, Baltimore, Maryland, United States; 9 Gladstone Insitute of Virology and Immunology, San Francisco, Calif, United States.


Journal of Infection | 2010

Acetylation of Tau Inhibits Its Degradation and Contributes to Tauopathy

Sang-Won Min; Seo-hyun Cho; Yungui Zhou; Sebastian Schroeder; Vahram Haroutunian; William W. Seeley; Eric J. Huang; Yong Shen; Eliezer Masliah; Chandrani Mukherjee; David J. Meyers; Philip A. Cole; Melanie Ott; Li Gan

Collaboration


Dive into the Sang-Won Min's collaboration.

Top Co-Authors

Avatar

Li Gan

University of California

View shared research outputs
Top Co-Authors

Avatar

Yungui Zhou

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chao Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Seo-hyun Cho

University of California

View shared research outputs
Top Co-Authors

Avatar

Yaqiao Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Eric J. Huang

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge