Network


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

Hotspot


Dive into the research topics where Yunjong Lee is active.

Publication


Featured researches published by Yunjong Lee.


Cell | 2011

PARIS (ZNF746) Repression of PGC-1α Contributes to Neurodegeneration in Parkinson's Disease

Joo Ho Shin; Han Seok Ko; Hochul Kang; Yunjong Lee; Yun Il Lee; Olga Pletinkova; Juan C. Troconso; Valina L. Dawson; Ted M. Dawson

A hallmark of Parkinsons disease (PD) is the preferential loss of substantia nigra dopamine neurons. Here, we identify a new parkin interacting substrate, PARIS (ZNF746), whose levels are regulated by the ubiquitin proteasome system via binding to and ubiquitination by the E3 ubiquitin ligase, parkin. PARIS is a KRAB and zinc finger protein that accumulates in models of parkin inactivation and in human PD brain. PARIS represses the expression of the transcriptional coactivator, PGC-1α and the PGC-1α target gene, NRF-1 by binding to insulin response sequences in the PGC-1α promoter. Conditional knockout of parkin in adult animals leads to progressive loss of dopamine (DA) neurons in a PARIS-dependent manner. Moreover, overexpression of PARIS leads to the selective loss of DA neurons in the substantia nigra, and this is reversed by either parkin or PGC-1α coexpression. The identification of PARIS provides a molecular mechanism for neurodegeneration due to parkin inactivation.


Cell | 1980

Receptor-mediated pinocytosis of mannose glycoconjugates by macrophages: Characterization and evidence for receptor recycling

Philip D. Stahl; Paul H. Schlesinger; Elaine Sigardson; Jane Somsel Rodman; Yunjong Lee

125I-Mannose--BSA is taken up by alveolar macrophages by receptor-mediated endocytosis. Uptake is macrophage-specific and does not occur in polymorphonuclear leukocytes. Binding (4 degrees C) and uptake (37 degrees C) of 125I--Man--BSA are time- and ligand concentration-dependent [Kuptake = 40 nM; Kd (4 degrees C) = 10 nM]. When adjusted for ligand degradation, ligand uptake is linear with time. Binding saturates at 60 min and requires Ca++. Following binding, ligand remains on the cell surface where it can be released by EGTA and trypsin. Internalization of prebound ligand occurs very rapidly (t 1/2 less than 5 min) when cells are warmed to 37 degrees C. Following internalization of prebound ligand, binding activity is rapidly recovered (t 1/2 less than 5 min). Trypsin treatment (4 degrees C) substantially reduces binding activity (greater than 70% per 30 min). However, binding activity is rapidly recovered in cells treated with trypsin at 4 degrees C by warming to 37 degrees C in the absence of added ligand. Trypsin treatment at 37 degrees C rapidly destroys binding and uptake. On the contrary, 4 degrees C trypsin treatment produces only a modest reduction in subsequent ligand uptake. These results, taken together with the observation that cycloheximide has no effect on ligand uptake, suggest that receptors must be spared from degradation and that reutilization of receptors probably occurs.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Phosphorylation by the c-Abl protein tyrosine kinase inhibits parkin's ubiquitination and protective function

Han Seok Ko; Yunjong Lee; Joo Ho Shin; Senthilkumar S. Karuppagounder; Bharathi Shrikanth Gadad; Anthony J. Koleske; Olga Pletnikova; Juan C. Troncoso; Valina L. Dawson; Ted M. Dawson

Mutations in PARK2/Parkin, which encodes a ubiquitin E3 ligase, cause autosomal recessive Parkinson disease (PD). Here we show that the nonreceptor tyrosine kinase c-Abl phosphorylates tyrosine 143 of parkin, inhibiting parkins ubiquitin E3 ligase activity and protective function. c-Abl is activated by dopaminergic stress and by dopaminergic neurotoxins, 1-methyl-4-phenylpyridinium (MPP+) in vitro and in vivo by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), leading to parkin inactivation, accumulation of the parkin substrates aminoacyl-tRNA synthetase-interacting multifunctional protein type 2 (AIMP2) (p38/JTV-1) and fuse-binding protein 1 (FBP1), and cell death. STI-571, a c-Abl-family kinase inhibitor, prevents the phosphorylation of parkin, maintaining parkin in a catalytically active and protective state. STI-571’s protective effects require parkin, as shRNA knockdown of parkin prevents STI-571 protection. Conditional knockout of c-Abl in the nervous system also prevents the phosphorylation of parkin, the accumulation of its substrates, and subsequent neurotoxicity in response to MPTP intoxication. In human postmortem PD brain, c-Abl is active, parkin is tyrosine-phosphorylated, and AIMP2 and FBP1 accumulate in the substantia nigra and striatum. Thus, tyrosine phosphorylation of parkin by c-Abl is a major posttranslational modification that inhibits parkin function, possibly contributing to pathogenesis of sporadic PD. Moreover, inhibition of c-Abl may be a neuroprotective approach in the treatment of PD.


Biochemical and Biophysical Research Communications | 1985

Novel approach to the study of the antigenicities and receptor functions of carbohydrate chains of glycoproteins

P.W. Tang; H.C. Gool; M. Hardy; Yunjong Lee; T. Felzi

This report describes the construction of neoglycolipids as a novel approach to determining the antigenicities and receptor functions of minute amounts of oligosaccharides derived from glycoproteins. Reduced oligosaccharides are converted into oligosaccharide alditols by controlled selective periodate oxidation and conjugated to phosphatidyl ethanolamine dipalmitoyl by reductive amination. The resulting neoglycolipids can be rendered multivalent by binding to polyvinylchloride or silica plates or they can be incorporated into liposomes and their antigenicities and receptor activities determined in low concentrations by direct binding or inhibition of binding assays. This approach, which has been successfully used with two monoclonal antibodies and a plant lectin, should be widely applicable to the direct analysis of O- and N-glycosidically linked carbohydrate chains of glycoproteins and proteoglycans both as antigens and recognition structures of diverse receptor systems.


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.


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.


Cold Spring Harbor Perspectives in Medicine | 2012

Animal Models of Parkinson's Disease: Vertebrate Genetics

Yunjong Lee; Valina L. Dawson; Ted M. Dawson

Parkinsons disease (PD) is a complex genetic disorder that is associated with environmental risk factors and aging. Vertebrate genetic models, especially mice, have aided the study of autosomal-dominant and autosomal-recessive PD. Mice are capable of showing a broad range of phenotypes and, coupled with their conserved genetic and anatomical structures, provide unparalleled molecular and pathological tools to model human disease. These models used in combination with aging and PD-associated toxins have expanded our understanding of PD pathogenesis. Attempts to refine PD animal models using conditional approaches have yielded in vivo nigrostriatal degeneration that is instructive in ordering pathogenic signaling and in developing therapeutic strategies to cure or halt the disease. Here, we provide an overview of the generation and characterization of transgenic and knockout mice used to study PD followed by a review of the molecular insights that have been gleaned from current PD mouse models. Finally, potential approaches to refine and improve current models are discussed.


Scientific Reports | 2015

The c-Abl inhibitor, Nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson's disease

Senthilkumar S. Karuppagounder; Saurav Brahmachari; Yunjong Lee; Valina L. Dawson; Ted M. Dawson; Hanseok Ko

c-Abl is activated in the brain of Parkinsons disease (PD) patients and in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice where it inhibits parkin through tyrosine phosphorylation leading to the accumulation of parkin substrates, and neuronal cell death. In the present study, we evaluated the in vivo efficacy of nilotinib, a brain penetrant c-Abl inhibitor, in the acute MPTP-induced model of PD. Our results show that administration of nilotinib reduces c-Abl activation and the levels of the parkin substrate, PARIS, resulting in prevention of dopamine (DA) neuron loss and behavioral deficits following MPTP intoxication. On the other hand, we observe no reduction in the tyrosine phosphorylation of parkin and the parkin substrate, AIMP2 suggesting that the protective effect of nilotinib may, in part, be parkin-independent or to the pharmacodynamics properties of nilotinib. This study provides a strong rationale for testing other brain permeable c-Abl inhibitors as potential therapeutic agents for the treatment of PD.


Cell Death and Disease | 2014

NLRP3 inflammasome activation by mitochondrial ROS in bronchial epithelial cells is required for allergic inflammation

Sehwan Kim; D I Kim; Sang-Hyun Kim; Hwa-Yong Lee; K S Lee; S H Cho; Yunjong Lee

Abnormality in mitochondria has been suggested to be associated with development of allergic airway disorders. In this study, to evaluate the relationship between mitochondrial reactive oxygen species (ROS) and NLRP3 inflammasome activation in allergic asthma, we used a newly developed mitochondrial ROS inhibitor, NecroX-5. NecroX-5 reduced the increase of mitochondrial ROS generation in airway inflammatory cells, as well as bronchial epithelial cells, NLRP3 inflammasome activation, the nuclear translocation of nuclear factor-κB, increased expression of various inflammatory mediators and pathophysiological features of allergic asthma in mice. Finally, blockade of IL-1β substantially reduced airway inflammation and hyperresponsiveness in the asthmatic mice. These findings suggest that mitochondrial ROS have a critical role in the pathogenesis of allergic airway inflammation through the modulation of NLRP3 inflammasome activation, providing a novel role of airway epithelial cells expressing NLRP3 inflammasome as an immune responder.


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.

Collaboration


Dive into the Yunjong Lee's collaboration.

Top Co-Authors

Avatar

Ted M. Dawson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Valina L. Dawson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

J.Y. Byeon

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Sun-Woo Kim

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Y.J. Lee

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Y.H. Kim

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Senthilkumar S. Karuppagounder

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sangwoo Ham

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar

Sun-Mee Lee

Sungkyunkwan University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge