Network


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

Hotspot


Dive into the research topics where Ho Chul Kang is active.

Publication


Featured researches published by Ho Chul Kang.


Science Signaling | 2011

Poly(ADP-Ribose) (PAR) Binding to Apoptosis-Inducing Factor Is Critical for PAR Polymerase-1–Dependent Cell Death (Parthanatos)

Yingfei Wang; No Soo Kim; Jean François Haince; Ho Chul Kang; Karen K. David; Shaida A. Andrabi; Guy G. Poirier; Valina L. Dawson; Ted M. Dawson

Poly(ADP-ribose) binds to apoptosis-inducing factor to trigger its release from mitochondria and induce cell death. Scoring a PAR on Death’s Course Overactivation of the DNA repair enzyme PARP-1 [poly(ADP-ribose) (PAR) polymerase-1] leads to “parthanatos,” a form of cell death that is distinct from apoptosis and necrosis and that depends on release of apoptosis-inducing factor (AIF) from mitochondria. Here, Wang et al. showed that PAR bound directly to AIF, disrupting AIF’s association with the mitochondria and allowing it to translocate to the nucleus to mediate cell death. Moreover, mutation of the PAR binding site of AIF enabled the authors to separate AIF’s role in parthanatos from its function in mitochondrial respiration. Identification of AIF as a PAR-binding protein could potentially lead to the development of compounds that inhibit this interaction, protecting against parthanatos, or compounds that mimic it and thereby promote parthanatos as an agent of death in malignant cells. The mitochondrial protein apoptosis-inducing factor (AIF) plays a pivotal role in poly(ADP-ribose) polymerase–1 (PARP-1)–mediated cell death (parthanatos), during which it is released from the mitochondria and translocates to the nucleus. We show that AIF is a high-affinity poly(ADP-ribose) (PAR)–binding protein and that PAR binding to AIF is required for parthanatos both in vitro and in vivo. AIF bound PAR at a site distinct from AIF’s DNA binding site, and this interaction triggered AIF release from the cytosolic side of the mitochondrial outer membrane. Mutation of the PAR binding site in AIF did not affect its NADH (reduced form of nicotinamide adenine dinucleotide) oxidase activity, its ability to bind FAD (flavin adenine dinucleotide) or DNA, or its ability to induce nuclear condensation. However, this AIF mutant was not released from mitochondria and did not translocate to the nucleus or mediate cell death after PARP-1 activation. These results suggest a mechanism for PARP-1 to initiate AIF-mediated cell death and indicate that AIF’s bioenergetic cell survival–promoting functions are separate from its effects as a mitochondrially derived death effector. Interference with the PAR-AIF interaction or PAR signaling may provide notable opportunities for preventing cell death after activation of PARP-1.


Cell | 2014

Ribosomal Protein s15 Phosphorylation Mediates LRRK2 Neurodegeneration in Parkinson’s Disease

Ian Martin; Jungwoo Wren Kim; Byoung Dae Lee; Ho Chul Kang; Jin Chong Xu; Hao Jia; Jeannette N. Stankowski; Min Sik Kim; Jun Zhong; Manoj Kumar; Shaida A. Andrabi; Yulan Xiong; Dennis W. Dickson; Zbigniew K. Wszolek; Akhilesh Pandey; Ted M. Dawson; Valina L. Dawson

Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinsons disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Iduna is a poly(ADP-ribose) (PAR)-dependent E3 ubiquitin ligase that regulates DNA damage

Ho Chul Kang; Yun Il Lee; Joo Ho Shin; Shaida A. Andrabi; Zhikai Chi; Jean Philippe Gagné; Yunjong Lee; Han Seok Ko; Byoung Dae Lee; Guy G. Poirier; Valina L. Dawson; Ted M. Dawson

Ubiquitin mediated protein degradation is crucial for regulation of cell signaling and protein quality control. Poly(ADP-ribose) (PAR) is a cell-signaling molecule that mediates changes in protein function through binding at PAR binding sites. Here we characterize the PAR binding protein, Iduna, and show that it is a PAR-dependent ubiquitin E3 ligase. Iduna’s E3 ligase activity requires PAR binding because point mutations at Y156A and R157A eliminate Iduna’s PAR binding and Iduna’s E3 ligase activity. Iduna’s E3 ligase activity also requires an intact really interesting new gene (RING) domain because Iduna possessing point mutations at either H54A or C60A is devoid of ubiquitination activity. Tandem affinity purification reveals that Iduna binds to a number of proteins that are either PARsylated or bind PAR including PAR polymerase-1, 2 (PARP1, 2), nucleolin, DNA ligase III, KU70, KU86, XRCC1, and histones. PAR binding to Iduna activates its E3 ligase function, and PAR binding is required for Iduna ubiquitination of PARP1, XRCC1, DNA ligase III, and KU70. Iduna’s PAR-dependent ubiquitination of PARP1 targets it for proteasomal degradation. Via PAR binding and ubiquitin E3 ligase activity, Iduna protects against cell death induced by the DNA damaging agent N-methyl-N-nitro-N-nitrosoguanidine (MNNG) and rescues cells from G1 arrest and promotes cell survival after γ-irradiation. Moreover, Iduna facilitates DNA repair by reducing apurinic/apyrimidinic (AP) sites after MNNG exposure and facilitates DNA repair following γ-irradiation as assessed by the comet assay. These results define Iduna as a PAR-dependent E3 ligase that regulates cell survival and DNA repair.


Science | 2016

Pathological α-synuclein transmission initiated by binding lymphocyte-activation gene 3

Xiaobo Mao; Michael Tianhao Ou; Senthilkumar S. Karuppagounder; Tae In Kam; Xiling Yin; Yulan Xiong; Preston Ge; George Essien Umanah; Saurav Brahmachari; Joo Ho Shin; Ho Chul Kang; Jianmin Zhang; Jinchong Xu; Rong Chen; Hyejin Park; Shaida A. Andrabi; Sung Ung Kang; Rafaella Araújo Gonçalves; Yu Liang; Shu Zhang; Chen Qi; Sharon Lam; James A. Keiler; Joel Tyson; Donghoon Kim; Nikhil Panicker; Seung Pil Yun; Creg J. Workman; Dario A. A. Vignali; Valina L. Dawson

INTRODUCTION Parkinson’s disease (PD) is the second most common neurodegenerative disorder and leads to slowness of movement, tremor, rigidity, and, in the later stages of PD, cognitive impairment. Pathologically, PD is characterized by the accumulation of α-synuclein in Lewy bodies and neurites. There is degeneration of neurons throughout the nervous system, with the degeneration of dopamine neurons in the substantia nigra pars compacta leading to the major symptoms of PD. RATIONALE In the brains of PD patients, pathologic α-synuclein seems to spread from cell to cell via self-amplification, propagation, and transmission in a stereotypical and topographical pattern among neighboring cells and/or anatomically connected brain regions. The spread or transmission of pathologic α-synuclein is emerging as a potentially important driver of PD pathogenesis. The underlying mechanisms and molecular entities responsible for the transmission of pathologic α-synuclein from cell to cell are not known, but the entry of pathologic α-synuclein into neurons is thought to occur, in part, through an active clathrin-dependent endocytic process. RESULTS Using recombinant α-synuclein preformed fibrils (PFF) as a model system with which to study the transmission of misfolded α-synuclein from neuron to neuron, we screened a library encoding transmembrane proteins for α-synuclein-biotin PFF–binding candidates via detection with streptavidin-AP (alkaline phosphatase) staining. Three positive clones were identified that bind α-synuclein PFF and include lymphocyte-activation gene 3 (LAG3), neurexin 1β, and amyloid β precursor-like protein 1 (APLP1). Of these three transmembrane proteins, LAG3 demonstrated the highest ratio of selectivity for α-synuclein PFF over the α-synuclein monomer. α-Synuclein PFF bind to LAG3 in a saturable manner (dissociation constant = 77 nM), whereas the α-synuclein monomer does not bind to LAG3. Co-immunoprecipitation also suggests that pathological α-synuclein PFF specifically bind to LAG3. Tau PFF, β-amyloid oligomer, and β-amyloid PFF do not bind to LAG3, indicating that LAG3 is specific for α-synuclein PFF. The internalization of α-synuclein PFF involves LAG3 because deletion of LAG3 reduces the endocytosis of α-synuclein PFF. LAG3 colocalizes with the endosomal guanosine triphosphatases Rab5 and Rab7 and coendocytoses with pathologic α-synuclein. Neuron-to-neuron transmission of pathologic α-synuclein and the accompanying pathology and neurotoxicity is substantially attenuated by deletion of LAG3 or by antibodies to LAG3. The lack of LAG3 also substantially delayed α-synuclein PFF–induced loss of dopamine neurons, as well as biochemical and behavioral deficits in vivo. CONCLUSION We discovered that pathologic α-synuclein transmission and toxicity is initiated by binding to LAG3 and that neuron-to-neuron transmission of pathological α-synuclein involves the endocytosis of exogenous α-synuclein PFF by the engagement of LAG3 on neurons. Depletion of LAG3 or antibodies to LAG3 substantially reduces the pathology set in motion by the transmission of pathologic α-synuclein. The identification of LAG3 as an α-synuclein PFF–binding protein provides a new target for developing therapeutics designed to slow the progression of PD and related α-synucleinopathies. LAG3 deletion or antibodies to LAG3 delay α-synuclein PFF transmission. Compared with wild-type neurons, binding and endocytosis of α-synuclein PFF is dramatically reduced with antibodies to LAG3 or when LAG3 is deleted, resulting in delayed pathologic α-synuclein transmission and toxicity. Illustration credit: I-Hsun Wu Emerging evidence indicates that the pathogenesis of Parkinson’s disease (PD) may be due to cell-to-cell transmission of misfolded preformed fibrils (PFF) of α-synuclein (α-syn). The mechanism by which α-syn PFF spreads from neuron to neuron is not known. Here, we show that LAG3 (lymphocyte-activation gene 3) binds α-syn PFF with high affinity (dissociation constant = 77 nanomolar), whereas the α-syn monomer exhibited minimal binding. α-Syn-biotin PFF binding to LAG3 initiated α-syn PFF endocytosis, transmission, and toxicity. Lack of LAG3 substantially delayed α-syn PFF–induced loss of dopamine neurons, as well as biochemical and behavioral deficits in vivo. The identification of LAG3 as a receptor that binds α-syn PFF provides a target for developing therapeutics designed to slow the progression of PD and related α-synucleinopathies.


Nature Medicine | 2011

Iduna protects the brain from glutamate excitotoxicity and stroke by interfering with poly(ADP-ribose) polymer-induced cell death

Shaida A. Andrabi; Ho Chul Kang; Jean François Haince; Yun Il Lee; Jian Zhang; Zhikai Chi; Andrew B. West; Raymond C. Koehler; Guy G. Poirier; Ted M. Dawson; Valina L. Dawson

Glutamate acting on N-methyl-D-aspartate (NMDA) receptors induces neuronal injury following stroke, through activation of poly(ADP-ribose) polymerase-1 (PARP-1) and generation of the death molecule poly(ADP-ribose) (PAR) polymer. Here we identify Iduna, a previously undescribed NMDA receptor–induced survival protein that is neuroprotective against glutamate NMDA receptor–mediated excitotoxicity both in vitro and in vivo and against stroke through interfering with PAR polymer–induced cell death (parthanatos). Idunas protective effects are independent and downstream of PARP-1 activity. Iduna is a PAR polymer–binding protein, and mutation at the PAR polymer binding site abolishes the PAR binding activity of Iduna and attenuates its protective actions. Iduna is protective in vivo against NMDA-induced excitotoxicity and middle cerebral artery occlusion–induced stroke in mice. To our knowledge, these results define Iduna as the first known endogenous inhibitor of parthanatos. Interfering with PAR polymer signaling could be a new therapeutic strategy for the treatment of neurologic disorders.


Nature Neuroscience | 2013

Parthanatos mediates AIMP2-activated age-dependent dopaminergic neuronal loss

Yunjong Lee; Senthilkumar S. Karuppagounder; Joo Ho Shin; Yun Il Lee; Hanseok Ko; Debbie Swing; Haisong Jiang; Sung Ung Kang; Byoung Dae Lee; Ho Chul Kang; Donghoon Kim; Lino Tessarollo; Valina L. Dawson; Ted M. Dawson

The defining pathogenic feature of Parkinsons disease is the age-dependent loss of dopaminergic neurons. Mutations and inactivation of parkin, an ubiquitin E3 ligase, induce Parkinsons disease through accumulation of pathogenic substrates. We found that transgenic overexpression of a parkin substrate, aminoacyl-tRNA synthetase complex interacting multifunctional protein-2 (AIMP2), led to a selective, age-dependent, progressive loss of dopaminergic neurons via activation of poly(ADP-ribose) polymerase-1 (PARP1). AIMP2 accumulation in vitro and in vivo resulted in PARP1 overactivation and dopaminergic cell toxicity via direct association of these proteins in the nucleus, providing a path to PARP1 activation other than DNA damage. Inhibition of PARP1 through gene deletion or drug inhibition reversed behavioral deficits and protected against dopamine neuron death in AIMP2 transgenic mice. These data indicate that brain-permeable PARP inhibitors could effectively delay or prevent disease progression in Parkinsons disease.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Parkin loss leads to PARIS-dependent declines in mitochondrial mass and respiration

Daniel A. Stevens; Yunjong Lee; Ho Chul Kang; Byoung Dae Lee; Yun Il Lee; Aaron Bower; Haisong Jiang; Sung Ung Kang; Shaida A. Andrabi; Valina L. Dawson; Joo Ho Shin; Ted M. Dawson

Significance Mutations or inactivation of parkin causes Parkinson’s disease (PD) in humans. Recent studies have focused on parkin’s role in mitochondrial quality control in the pathogenesis of PD, including defects in mitophagy, mitochondrial fission, fusion, and transport. This study shows that parkin also controls mitochondrial biogenesis and that defects in mitochondrial biogenesis drive the loss of dopamine (DA) neurons due to the absence of parkin. The findings support the role of parkin in regulating multiple arms of mitochondrial quality control and suggest that maintaining mitochondrial biogenesis is critically important in the survival of DA neurons. Mutations in parkin lead to early-onset autosomal recessive Parkinson’s disease (PD) and inactivation of parkin is thought to contribute to sporadic PD. Adult knockout of parkin in the ventral midbrain of mice leads to an age-dependent loss of dopamine neurons that is dependent on the accumulation of parkin interacting substrate (PARIS), zinc finger protein 746 (ZNF746), and its transcriptional repression of PGC-1α. Here we show that adult knockout of parkin in mouse ventral midbrain leads to decreases in mitochondrial size, number, and protein markers consistent with a defect in mitochondrial biogenesis. This decrease in mitochondrial mass is prevented by short hairpin RNA knockdown of PARIS. PARIS overexpression in mouse ventral midbrain leads to decreases in mitochondrial number and protein markers and PGC-1α–dependent deficits in mitochondrial respiration. Taken together, these results suggest that parkin loss impairs mitochondrial biogenesis, leading to declining function of the mitochondrial pool and cell death.


Cell | 2011

The AAA+ ATPase Thorase Regulates AMPA Receptor-Dependent Synaptic Plasticity and Behavior

Jianmin Zhang; Yue Wang; Zhikai Chi; Matthew J. Keuss; Ying Min Emily Pai; Ho Chul Kang; Joo Ho Shin; Artem Bugayenko; Hong Wang; Yulan Xiong; Mikhail V. Pletnikov; Mark P. Mattson; Ted M. Dawson; Valina L. Dawson

The synaptic insertion or removal of AMPA receptors (AMPAR) plays critical roles in the regulation of synaptic activity reflected in the expression of long-term potentiation (LTP) and long-term depression (LTD). The cellular events underlying this important process in learning and memory are still being revealed. Here we describe and characterize the AAA+ ATPase Thorase, which regulates the expression of surface AMPAR. In an ATPase-dependent manner Thorase mediates the internalization of AMPAR by disassembling the AMPAR-GRIP1 complex. Following genetic deletion of Thorase, the internalization of AMPAR is substantially reduced, leading to increased amplitudes of miniature excitatory postsynaptic currents, enhancement of LTP, and elimination of LTD. These molecular events are expressed as deficits in learning and memory in Thorase null mice. This study identifies an AAA+ ATPase that plays a critical role in regulating the surface expression of AMPAR and thereby regulates synaptic plasticity and learning and memory.


Molecular & Cellular Proteomics | 2014

Protein Microarray Characterization of the S-Nitrosoproteome

Yun Il Lee; Daniel Giovinazzo; Ho Chul Kang; Yunjong Lee; Jun Seop Jeong; Paschalis-Thomas Doulias; Zhi Xie; Jianfei Hu; Mehdi Ghasemi; Harry Ischiropoulos; Jiang Qian; Heng Zhu; Seth Blackshaw; Valina L. Dawson; Ted M. Dawson

Nitric oxide (NO) mediates a substantial part of its physiologic functions via S-nitrosylation, however the cellular substrates for NO-mediated S-nitrosylation are largely unknown. Here we describe the S-nitrosoproteome using a high-density protein microarray chip containing 16,368 unique human proteins. We identified 834 potentially S-nitrosylated human proteins. Using a unique and highly specific labeling and affinity capture of S-nitrosylated proteins, 138 cysteine residues on 131 peptides in 95 proteins were determined, defining critical sites of NOs actions. Of these cysteine residues 113 are novel sites of S-nitrosylation. A consensus sequence motif from these 834 proteins for S-nitrosylation was identified, suggesting that the residues flanking the S-nitrosylated cysteine are likely to be the critical determinant of whether the cysteine is S-nitrosylated. We identify eight ubiquitin E3 ligases, RNF10, RNF11, RNF41, RNF141, RNF181, RNF208, WWP2, and UBE3A, whose activities are modulated by S-nitrosylation, providing a unique regulatory mechanism of the ubiquitin proteasome system. These results define a new and extensive set of proteins that are susceptible to NO regulation via S-nitrosylation. Similar approaches could be used to identify other post-translational modification proteomes.


Nature Communications | 2015

LRRK2 G2019S mutation attenuates microglial motility by inhibiting focal adhesion kinase

Insup Choi; Beomsue Kim; Ji-Won Byun; Sung Hoon Baik; Yun Hyun Huh; Jong-hyeon Kim; Inhee Mook-Jung; Woo Keun Song; Joo-Ho Shin; Hyemyung Seo; Young Ho Suh; Ilo Jou; Sang Myun Park; Ho Chul Kang; Eun-hye Joe

In response to brain injury, microglia rapidly extend processes that isolate lesion sites and protect the brain from further injury. Here we report that microglia carrying a pathogenic mutation in the Parkinsons disease (PD)-associated gene, G2019S-LRRK2 (GS-Tg microglia), show retarded ADP-induced motility and delayed isolation of injury, compared with non-Tg microglia. Conversely, LRRK2 knockdown microglia are highly motile compared with control cells. In our functional assays, LRRK2 binds to focal adhesion kinase (FAK) and phosphorylates its Thr–X–Arg/Lys (TXR/K) motif(s), eventually attenuating FAK activity marked by decreased pY397 phosphorylation (pY397). GS-LRRK2 decreases the levels of pY397 in the brain, microglia and HEK cells. In addition, treatment with an inhibitor of LRRK2 kinase restores pY397 levels, decreased pTXR levels and rescued motility of GS-Tg microglia. These results collectively suggest that G2019S mutation of LRRK2 may contribute to the development of PD by inhibiting microglial response to brain injury.

Collaboration


Dive into the Ho Chul Kang's collaboration.

Top Co-Authors

Avatar

Valina L. Dawson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ted M. Dawson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shaida A. Andrabi

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yunjong Lee

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Joo Ho Shin

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Yun Il Lee

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Donghoon Kim

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Haisong Jiang

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Senthilkumar S. Karuppagounder

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge