Network


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

Hotspot


Dive into the research topics where Jung A. Woo is active.

Publication


Featured researches published by Jung A. Woo.


Cell Death & Differentiation | 2012

Pivotal role of the RanBP9-cofilin pathway in Aβ-induced apoptosis and neurodegeneration.

Jung A. Woo; A-Rong Jung; Madepalli K. Lakshmana; A Bedrossian; Yeory Lim; Jung Hyun Bu; S A Park; Edward H. Koo; Inhee Mook-Jung; David E. Kang

Neurodegeneration associated with amyloid β (Aβ) peptide accumulation, synaptic loss, neuroinflammation, tauopathy, and memory impairments encompass the pathophysiological features of Alzheimers disease (AD). We previously reported that the scaffolding protein RanBP9, which is overall increased in brains of AD patients, simultaneously promotes Aβ generation and focal adhesion disruption by accelerating the endocytosis of amyloid precursor protein (APP) and β1-integrin, respectively. Here, we show that RanBP9 protein levels are increased by fourfold in FAD mutant APP transgenic mice. Accordingly, RanBP9 transgenic mice demonstrate significantly increased synapse loss, neurodegeneration, gliosis, and spatial memory deficits. RanBP9 overexpression promotes apoptosis and potentiates Aβ-induced neurotoxicity independent of its capacity to promote Aβ generation. Conversely, RanBP9 reduction by siRNA or gene dosage mitigates Aβ-induced neurotoxicity. Importantly, RanBP9 activates/dephosphorylates cofilin, a key regulator of actin dynamics and mitochondria-mediated apoptosis, and siRNA knockdown of cofilin abolishes both Aβ and RanBP9-induced apoptosis. These findings implicate the RanBP9–cofilin pathway as critical therapeutic targets not only for stemming Aβ generation but also antagonizing Aβ-induced neurotoxicity.


Cell Death and Disease | 2013

Cooperative role of RanBP9 and P73 in mitochondria-mediated apoptosis

Tian Liu; Seung-Eon Roh; Jung A. Woo; Hoon Ryu; David E. Kang

Mitochondrial dysfunction and synaptic damage are critical early features of Alzheimer’s disease (AD) associated with amyloid β (Aβ) and τ. We previously reported that the scaffolding protein RanBP9, which is overall increased in AD, simultaneously promotes Aβ generation and focal adhesion disruption by accelerating the endocytosis of APP and β1-integrin, respectively. Moreover, RanBP9 induces neurodegeneration in vitro and in vivo and mediates Aβ-induced neurotoxicity. However, little is known regarding the mechanisms underlying such neurotoxic processes. Here, we show that RanBP9 induces the loss of mitochondrial membrane potential and increase in mitochondrial superoxides associated with decrease in Bcl-2, increase in Bax protein and oligomerization, fragmentation of mitochondria, and cytochrome c release. RanBP9-induced neurotoxic changes are significantly prevented by the mitochondrial fission inhibitor Mdivi-1 and by classical inhibitors of the mitochondrial apoptosis, XIAP, Bcl-2, and Bcl-xl. RanBP9 physically interacts with the tumor suppressor p73 and increases endogenous p73α levels at both transcriptional and post-translational levels;moreover, the knockdown of endogenous p73 by siRNA effectively blocks RanBP9 and Aβ1-42-induced mitochondria-mediated cell death. Conversely, siRNA knockdown of endogenous RanBP9 also suppresses p73-induced apoptosis, suggesting that RanBP9 and p73 have cooperative roles in inducing cell death. Taken together, these finding implicate the RanBP9/p73 complex in mitochondria-mediated apoptosis in addition to its role in enhancing Aβ generation.


The FASEB Journal | 2012

Pivotal role of RanBP9 in integrin-dependent focal adhesion signaling and assembly

Jung A. Woo; Seung-Eon Roh; Madepalli K. Lakshmana; David E. Kang

Accumulation of the amyloid β (Aβ) peptide derived from the amyloid precursor protein (APP) plays a central role in the pathogenesis of Alzheimers disease (AD). We previously reported that the scaffolding protein RanBP9 is markedly increased in AD brains and promotes Aβ generation by scaffolding APP/BACE1/LRP complexes together and accelerating APP endocytosis. Because APP, LRP, and RanBP9 all physically interact with β‐integrins, we investigated whether RanBP9 alters integrin‐dependent cell adhesion and focal adhesion signaling. Here, we show that RanBP9 overexpression dramatically disrupts integrindependent∗∗∗ cell attachment and spreading in NIH3T3 and hippocampus‐derived HT22 cells, concomitant with strongly decreased Pyk2/paxillin signaling and talin/vinculin localization in focal adhesion complexes. Conversely, RanBP9 knockdown robustly promotes cell attachment, spreading, and focal adhesion signaling and assembly. Cell surface biotinylation and endocytosis assays reveal that RanBP9 overexpression and RanBP9 siRNA potently reduces and increases surface β1‐integrin and LRP by accelerating and inhibiting their endocytosis, respectively. Primary hippocampal neurons derived from RanBP9‐transgenic mice also demonstrate severely reduced levels of surface β1‐integrin, LRP, and APP, as well as neurite arborization. Therefore, these data indicate that RanBP9 simultaneously inhibits cell‐adhesive processes and enhances Aβ generation by accelerating APP, LRP, and β1‐integrin endocytosis.—Woo, J. A., Roh, S.‐E., Lakshmana, M. K., Kang, D. E. Pivotal role of RanBP9 in integrin‐dependent focal adhesion signaling and assembly. FASEB J. 26, 1672‐1681 (2012). www.fasebj.org


Cell Death and Disease | 2015

RanBP9 at the intersection between cofilin and Aβ pathologies: rescue of neurodegenerative changes by RanBP9 reduction

Jung A. Woo; Taylor Boggess; Courtney Uhlar; Xinming Wang; Hirah Khan; George Cappos; Aurelie Joly-Amado; E De Narvaez; S Majid; L S Minamide; J R Bamburg; Dave Morgan; Edwin J. Weeber; David E. Kang

Molecular pathways underlying the neurotoxicity and production of amyloid β protein (Aβ) represent potentially promising therapeutic targets for Alzheimer’s disease (AD). We recently found that overexpression of the scaffolding protein RanBP9 increases Aβ production in cell lines and in transgenic mice while promoting cofilin activation and mitochondrial dysfunction. Translocation of cofilin to mitochondria and induction of cofilin–actin pathology require the activation/dephosphorylation of cofilin by Slingshot homolog 1 (SSH1) and cysteine oxidation of cofilin. In this study, we found that endogenous RanBP9 positively regulates SSH1 levels and mediates Aβ-induced translocation of cofilin to mitochondria and induction of cofilin–actin pathology in cultured cells, primary neurons, and in vivo. Endogenous level of RanBP9 was also required for Aβ-induced collapse of growth cones in immature neurons (days in vitro 9 (DIV9)) and depletion of synaptic proteins in mature neurons (DIV21). In vivo, amyloid precursor protein (APP)/presenilin-1 (PS1) mice exhibited 3.5-fold increased RanBP9 levels, and RanBP9 reduction protected against cofilin–actin pathology, synaptic damage, gliosis, and Aβ accumulation associated with APP/PS1 mice. Brains slices derived from APP/PS1 mice showed significantly impaired long-term potentiation (LTP), and RanBP9 reduction significantly enhanced paired pulse facilitation and LTP, as well as partially rescued contextual memory deficits associated with APP/PS1 mice. Therefore, these results underscore the critical importance of endogenous RanBP9 not only in Aβ accumulation but also in mediating the neurotoxic actions of Aβ at the level of synaptic plasticity, mitochondria, and cofilin–actin pathology via control of the SSH1-cofilin pathway in vivo.


Cell Death & Differentiation | 2015

Slingshot-Cofilin activation mediates mitochondrial and synaptic dysfunction via Aβ ligation to β1-integrin conformers.

Jung A. Woo; Xingyu Zhao; Hirah Khan; C Penn; Xinming Wang; Aurelie Joly-Amado; Edwin J. Weeber; Dave Morgan; David E. Kang

The accumulation of amyloid-β protein (Aβ) is an early event associated with synaptic and mitochondrial damage in Alzheimer’s disease (AD). Recent studies have implicated the filamentous actin (F-actin) severing protein, Cofilin, in synaptic remodeling, mitochondrial dysfunction, and AD pathogenesis. However, whether Cofilin is an essential component of the AD pathogenic process and how Aβ impinges its signals to Cofilin from the neuronal surface are unknown. In this study, we found that Aβ42 oligomers (Aβ42O, amyloid-β protein 1–42 oligomers) bind with high affinity to low or intermediate activation conformers of β1-integrin, resulting in the loss of surface β1-integrin and activation of Cofilin via Slingshot homology-1 (SSH1) activation. Specifically, conditional loss of β1-integrin prevented Aβ42O-induced Cofilin activation, and allosteric modulation or activation of β1-integrin significantly reduced Aβ42O binding to neurons while blocking Aβ42O-induced reactive oxygen species (ROS) production, mitochondrial dysfunction, depletion of F-actin/focal Vinculin, and apoptosis. Cofilin, in turn, was required for Aβ42O-induced loss of cell surface β1-integrin, disruption of F-actin/focal Talin–Vinculin, and depletion of F-actin-associated postsynaptic proteins. SSH1 reduction, which mitigated Cofilin activation, prevented Aβ42O-induced mitochondrial Cofilin translocation and apoptosis, while AD brain mitochondria contained significantly increased activated/oxidized Cofilin. In mechanistic support in vivo, AD mouse model (APP (amyloid precursor protein)/PS1) brains contained increased SSH1/Cofilin and decreased SSH1/14-3-3 complexes, indicative of SSH1–Cofilin activation via release of SSH1 from 14-3-3. Finally, genetic reduction in Cofilin rescued APP/Aβ-induced synaptic protein loss and gliosis in vivo as well as deficits in long-term potentiation (LTP) and contextual memory in APP/PS1 mice. These novel findings therefore implicate the essential involvement of the β1-integrin–SSH1–Cofilin pathway in mitochondrial and synaptic dysfunction in AD.


The FASEB Journal | 2013

Mitochondrial dysfunction and calcium deregulation by the RanBP9-cofilin pathway

Seung-Eon Roh; Jung A. Woo; Madepalli K. Lakshmana; Courtney Uhlar; Vinishaa Ankala; Taylor Boggess; Tian Liu; Yun-Hwa Hong; Inhee Mook-Jung; Sang Jeong Kim; David E. Kang

Mitochondrial dysfunction and synaptic damage are important features of Alzheimers disease (AD) associated with amyloid β (Aβ) and tau. We reported previously that the scaffolding protein RanBP9, which is overall increased in brains of patients with AD and in mutant APP transgenic mice, simultaneously promotes Aβ generation and focal adhesion disruption by accelerating the endocytosis of APP and β1‐integrin, respectively. Moreover, RanBP9 induces neurodegeneration in vitro and in vivo and mediates Aβ‐induced neurotoxicity. Here we show in primary hippocampal neurons that RanBP9 potentiates Aβ‐induced reactive oxygen species (ROS) overproduction, apoptosis, and calcium deregulation. Analyses of calcium‐handling measures demonstrate that RanBP9 selectively delays the clearance of cytosolic Ca2+ mediated by the mitochondrial calcium uniporter through a process involving the translocation of cofilin into mitochondria and oxidative mechanisms. Further, RanBP9 retards the anterograde axonal transport of mitochondria in primary neurons and decreases synaptic mitochondrial activity in brain. These data indicate that RanBP9, cofilin, and Aβ mimic and potentiate each other to produce mitochondrial dysfunction, ROS overproduction, and calcium deregulation, which leads to neurodegenerative changes reminiscent of those seen in AD.—Roh. S.‐E., Woo, J. A., Lakshmana, M. K., Uhlar, C., Ankala, V., Boggess, T., Liu, T., Hong, Y.‐H., Mook‐Jung, I., Kim, S. J., Kang, D. E., Mitochondrial dysfunction and calcium deregulation by the RanBP9‐cofilin pathway. FASEB J. 27, 4776–4789 (2013). www.fasebj.org


Nature Communications | 2017

Loss of function CHCHD10 mutations in cytoplasmic TDP-43 accumulation and synaptic integrity

Jung A. Woo; Tian Liu; Courtney Trotter; Cenxiao C. Fang; Emillio De Narvaez; Patrick LePochat; Drew Maslar; Anusha Bukhari; Xingyu Zhao; Andrew Deonarine; Sandy D. Westerheide; David E. Kang

Although multiple CHCHD10 mutations are associated with the spectrum of familial and sporadic frontotemporal dementia–amyotrophic lateral sclerosis (FTD–ALS) diseases, neither the normal function of endogenous CHCHD10 nor its role in the pathological milieu (that is, TDP-43 pathology) of FTD/ALS have been investigated. In this study, we made a series of observations utilizing Caenorhabditis elegans models, mammalian cell lines, primary neurons and mouse brains, demonstrating that CHCHD10 normally exerts a protective role in mitochondrial and synaptic integrity as well as in the retention of nuclear TDP-43, whereas FTD/ALS-associated mutations (R15L and S59L) exhibit loss of function phenotypes in C. elegans genetic complementation assays and dominant negative activities in mammalian systems, resulting in mitochondrial/synaptic damage and cytoplasmic TDP-43 accumulation. As such, our results provide a pathological link between CHCHD10-associated mitochondrial/synaptic dysfunction and cytoplasmic TDP-43 inclusions.


Experimental Neurobiology | 2011

The Interface between Cytoskeletal Aberrations and Mitochondrial Dysfunction in Alzheimer's Disease and Related Disorders

David E. Kang; Seung Eon Roh; Jung A. Woo; Tian Liu; Jung Hyun Bu; A-Rong Jung; Yeory Lim

The major defining pathological hallmarks of Alzheimers disease (AD) are the accumulations of Aβ in senile plaques and hyperphosphorylated tau in neurofibrillary tangles and neuropil threads. Recent studies indicate that rather than these insoluble lesions, the soluble Aβ oligomers and hyperphosphorylated tau are the toxic agents of AD pathology. Such pathological protein species are accompanied by cytoskeletal changes, mitochondrial dysfunction, Ca2+ dysregulation, and oxidative stress. In this review, we discuss how the binding of Aβ to various integrins, defects in downstream focal adhesion signaling, and activation of cofilin can impact mitochondrial dysfunction, cytoskeletal changes, and tau pathology induced by Aβ oligomers. Such pathological consequences can also feedback to further activate cofilin to promote cofilin pathology. We also suggest that the mechanism of Aβ generation by the endocytosis of APP is mechanistically linked with perturbations in integrin-based focal adhesion signaling, as APP, LRP, and β-integrins are physically associated with each other.


Human Molecular Genetics | 2017

Enhanced tau pathology via RanBP9 and Hsp90/Hsc70 chaperone complexes

Jung A. Woo; Tian Liu; Xingyu Zhao; Courtney Trotter; Ksenia Yrigoin; Sara Cazzaro; Emilio De Narvaez; Hirah Khan; Richard Witas; Anusha Bukhari; Kamal Makati; Xinming Wang; Chad A. Dickey; David E. Kang

&NA; Accumulation of amyloid &bgr; (A&bgr;) and tau represent the two major pathological hallmarks of Alzheimers disease (AD). Despite the critical importance of A&bgr; accumulation as an early event in AD pathogenesis, multiple lines of evidence indicate that tau is required to mediate A&bgr;‐induced neurotoxic signals in neurons. We have previously shown that the scaffolding protein Ran‐binding protein 9 (RanBP9), which is highly elevated in brains of AD and AD mouse models, both enhances A&bgr; production and mediates A&bgr;‐induced neurotoxicity. However, it is unknown whether and how RanBP9 transmits A&bgr;‐induced neurotoxic signals to tau. Here we show for the first time that overexpression or knockdown of RanBP9 directly enhances and reduces tau levels, respectively, in vitro and in vivo. Such changes in tau levels are associated with the ability of RanBP9 to physically interact with tau and heat shock protein 90/heat shock cognate 70 (Hsp90/Hsc70) complexes. Meanwhile, both RanBP9 and tau levels are simultaneously reduced by Hsp90 or Hsc70 inhibitors, whereas overexpression or knockdown of RanBP9 significantly diminishes the anti‐tau potency of Hsp90/Hsc70 inhibitors as well as Hsc70 variants (WT & E175S). Further, RanBP9 increases the capacity for Hsp90 and Hsc70 complexes to bind ATP and enhances their ATPase activities in vitro. These observations in vitro and cell lines are recapitulated in primary neurons and in vivo, as genetic reduction in RanBP9 not only ameliorates tauopathy in Tau‐P301S mice but also rescues the deficits in synaptic integrity and plasticity.


Scientific Reports | 2017

Cofilin-mediated Neuronal Apoptosis via p53 Translocation and PLD1 Regulation

Tian Liu; Fang Wang; Patrick LePochat; Jung A. Woo; Mohammed Zaheen Bukhari; Kyung Woo Hong; Courtney Trotter; David E. Kang

Amyloid β (Aβ) accumulation is an early event in the pathogenesis of Alzheimer’s disease (AD), leading to mitochondrial and synaptic dysfunction, tau accumulation, and eventual neuronal death. While the p53 apoptotic pathway has clearly been associated with Aβ deposits and neuronal apoptosis, the critical upstream factors contributing to p53 activation in AD are not well understood. We have previously shown that cofilin activation plays a pivotal role in Aβ-induced mitochondrial and synaptic dysfunction. In this study, we show that activated cofilin (S3A) preferentially forms a complex with p53 and promotes its mitochondrial and nuclear localization, resulting in transcription of p53-responsive genes and promotion of apoptosis. Conversely, reduction of endogenous cofilin by knockdown or genetic deficiency inhibits mitochondrial and nuclear translocation of p53 in cultured cells and in APP/PS1 mice. This cofilin-p53 pro-apoptotic pathway is subject to negative regulation by PLD1 thorough cofilin inactivation and inhibition of cofilin/p53 complex formation. Finally, activated cofilin is unable to induce apoptosis in cells genetically lacking p53. These findings taken together indicate that cofilin coopts and requires the nuclear and mitochondrial pro-apoptotic p53 program to induce and execute apoptosis, while PLD1 functions in a regulatory multi-brake capacity in this pathway.

Collaboration


Dive into the Jung A. Woo's collaboration.

Top Co-Authors

Avatar

David E. Kang

University of California

View shared research outputs
Top Co-Authors

Avatar

Tian Liu

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Seung-Eon Roh

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Hirah Khan

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Madepalli K. Lakshmana

Torrey Pines Institute for Molecular Studies

View shared research outputs
Top Co-Authors

Avatar

A-Rong Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Inhee Mook-Jung

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Yeory Lim

Seoul National University

View shared research outputs
Top Co-Authors

Avatar

Aurelie Joly-Amado

University of South Florida

View shared research outputs
Top Co-Authors

Avatar

Courtney Trotter

University of South Florida

View shared research outputs
Researchain Logo
Decentralizing Knowledge