18 results on '"Yoonji Lee"'
Search Results
2. Adaptable Small Ligand of CYP1 Enzymes for Use in Understanding the Structural Features Determining Isoform Selectivity
- Author
-
Sanghee Kim, Sun Choi, Yoonji Lee, Sundarapandian Thangapandian, Joo Youn Lee, Hyunkyung Cho, Young-Jin Chun, and Chaemin Lim
- Subjects
Gene isoform ,chemistry.chemical_classification ,biology ,Stereochemistry ,Organic Chemistry ,Cytochrome P450 ,Stilbenoid ,Biochemistry ,Hydrophobic effect ,Molecular dynamics ,Enzyme ,chemistry ,Docking (molecular) ,Drug Discovery ,biology.protein ,Selectivity - Abstract
[Image: see text] Although several families of compounds have been identified as scaffolds for inhibitors of the CYP1 family, the isoform selectivity determining structural features has not been fully clarified at the molecular interaction level. We studied the CYP1 isoform selectivity for stilbenoid inhibitors using integrated induced fit docking and molecular dynamics simulations. The hydrophobic interactions with the specific phenylalanine residues in the F helix are correlated with inhibitory potency in the CYP1 family. Through this study, we found that the adaptable, small, and semirigid ligand is a promising starting point for the development of isoform-selective inhibitors and investigation of selectivity-determining features.
- Published
- 2018
3. pCRM1exportome: database of predicted CRM1-dependent Nuclear Export Signal (NES) motifs in cancer-related genes
- Author
-
Yoonji Lee, Jimin Pei, Nick V. Grishin, Yuh Min Chook, and Jordan M. Baumhardt
- Subjects
Statistics and Probability ,viruses ,Active Transport, Cell Nucleus ,Receptors, Cytoplasmic and Nuclear ,Biology ,Karyopherins ,computer.software_genre ,Biochemistry ,environment and public health ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Humans ,Nuclear export signal ,Molecular Biology ,030304 developmental biology ,Supplementary data ,Cell Nucleus ,Nuclear Export Signals ,0303 health sciences ,COSMIC cancer database ,Database ,Reproducibility of Results ,Computer Science Applications ,Computational Mathematics ,Cancer related genes ,Applications Note ,Computational Theory and Mathematics ,030220 oncology & carcinogenesis ,Proteome ,Cancer gene ,lipids (amino acids, peptides, and proteins) ,UniProt ,Sequence motif ,computer - Abstract
Motivation The consensus pattern of Nuclear Export Signal (NES) is a short sequence motif that is commonly identified in protein sequences, whether the motif acts as an NES (true positive) or not (false positive). Finding more plausible NES functioning regions among the vast array of consensus-matching segments would provide an interesting resource for further experimental validation. Better defined NES should also allow meaningful mapping of cancer-related mutation positions, leading to plausible explanations for the relationship between nuclear export and disease. Results Possible NES candidate regions are extracted from the cancer-related human reference proteome. Extracted NES are scored for reliability by combining sequence-based and structure-based approaches. The confidently identified NES candidate motifs were checked for overlap with cancer-related mutation positions annotated in the COSMIC database. Among the ∼700 cancer-related sequences in the COSMIC Cancer Gene Census, 178 sequences are predicted to have possible NES motifs containing cancer-related mutations at their key positions. These lists are organized into our database (pCRM1exportome), and other protein sequences in the human reference proteome can also be retrieved by their UniProt IDs. Availability and implementation The database is freely available at http://prodata.swmed.edu/pCRM1exportome. Supplementary information Supplementary data are available at Bioinformatics online.
- Published
- 2019
4. In Vivo Albumin Traps Photosensitizer Monomers from Self-Assembled Phthalocyanine Nanovesicles: A Facile and Switchable Theranostic Approach
- Author
-
Nahyun Kwon, Yoonji Lee, Ki Taek Nam, Gyoungmi Kim, Su Cheong Yeom, Juyoung Yoon, Yejin Cho, Sun Choi, Dayoung Lee, Jian-Dong Huang, Sungsook Yu, Tian Guo, and Xingshu Li
- Subjects
Male ,Fluorescence-lifetime imaging microscopy ,Indoles ,medicine.medical_treatment ,Photodynamic therapy ,Mice, Transgenic ,Plasma protein binding ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Catalysis ,Theranostic Nanomedicine ,Colloid and Surface Chemistry ,In vivo ,Pregnancy ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Photosensitizer ,Serum Albumin ,Fluorescent Dyes ,Nanocomposite ,Photosensitizing Agents ,Chemistry ,Albumin ,General Chemistry ,Xenograft Model Antitumor Assays ,0104 chemical sciences ,Photochemotherapy ,Drug delivery ,Biophysics ,Nanoparticles ,Female ,Protein Binding - Abstract
Albumin is a promising candidate as a biomarker for potential disease diagnostics and has been extensively used as a drug delivery carrier for decades. In these two directions, many albumin-detecting probes and exogenous albumin-based nanocomposite delivery systems have been developed. However, there are only a few cases demonstrating the specific interactions of exogenous probes with albumin in vivo, and nanocomposite delivery systems usually suffer from tedious fabrication processes and potential toxicity of the complexes. Herein, we demonstrate a facile "one-for-all" switchable nanotheranostic (NanoPcS) for both albumin detection and cancer treatment. In particular, the in vivo specific binding between albumin and PcS, arising from the disassembly of injected NanoPcS, is confirmed using an inducible transgenic mouse system. Fluorescence imaging and antitumor tests on different tumor models suggest that NanoPcS has superior tumor-targeting ability and the potential for time-modulated, activatable photodynamic therapy.
- Published
- 2018
5. Structure–Activity Relationships of Neplanocin A Analogues as S-Adenosylhomocysteine Hydrolase Inhibitors and Their Antiviral and Antitumor Activities
- Author
-
Sun Choi, Lak Shin Jeong, Tong Shin Chang, Varughese A. Mulamoottil, Minsoo Noh, Kawon Oh, Akshata Nayak, Pramod K. Sahu, Yoonji Lee, Yang Won Moon, Ji Yong Jang, Gyudong Kim, Sang Kook Lee, Girish Chandra, and Jayoung Song
- Subjects
Adenosine ,Stereochemistry ,Antineoplastic Agents ,Inhibitory postsynaptic potential ,Antiviral Agents ,Structure-Activity Relationship ,Cell Line, Tumor ,Neoplasms ,Chlorocebus aethiops ,Drug Discovery ,Hydrolase ,Animals ,Humans ,Structure–activity relationship ,Moiety ,Enzyme Inhibitors ,Vero Cells ,IC50 ,chemistry.chemical_classification ,biology ,Chemistry ,Adenine ,Adenosylhomocysteinase ,Active site ,Molecular Docking Simulation ,Enzyme ,Biochemistry ,Virus Diseases ,Viruses ,biology.protein ,Molecular Medicine ,Pharmacophore - Abstract
On the basis of the potent inhibitory activity of neplanocin A (1) against S-adenosylhomocysteine (AdoHcy) hydrolase, we analyzed the comprehensive structure-activity relationships by modifying the adenine and carbasugar moiety of 1 to find the pharmacophore in the active site of the enzyme. The introduction of 7-deazaadenine instead of adenine eliminated the inhibitory activity against the AdoHcy hydrolase, while 3-deazaadenine maintained the inhibitory activity of the enzyme, indicating that N-7 is essential for its role as a hydrogen bonding acceptor. The substitution of hydrogen at the 6'-position with fluorine increased the inhibitory activity of the enzyme. The one-carbon homologation at the 5'-position generally decreased the inhibitory activity of the enzyme, indicating that steric repulsion exists. A molecular docking study also supported these experimental data. In this study, 6'-fluoroneplanocin A (2) was the most potent inhibitor of AdoHcy hydrolase (IC50 = 0.24 μM). It showed a potent anti-VSV activity (EC50 = 0.43 μM) and potent anticancer activity in all the human tumor cell lines tested.
- Published
- 2015
6. Anti-angiogenic activity of thienopyridine derivative LCB03-0110 by targeting VEGFR-2 and JAK/STAT3 Signalling
- Author
-
Minghua Cui, Sun Choi, Sun-Young Kim, Hyun Yoo, S.-K. Lee, Yoonji Lee, Beom-Seok Yang, Byung Hak Kim, Jong Un Cho, Young Guen Kwon, Hyang Sook Lee, and Tae-Yoon Kim
- Subjects
Male ,Models, Molecular ,STAT3 Transcription Factor ,Lung Neoplasms ,Cell Survival ,Angiogenesis ,Aminopyridines ,Mice, Nude ,Angiogenesis Inhibitors ,Thiophenes ,Dermatology ,Biology ,Biochemistry ,Protein Structure, Secondary ,CSK Tyrosine-Protein Kinase ,Mice ,chemistry.chemical_compound ,Adenosine Triphosphate ,Cell Line, Tumor ,Human Umbilical Vein Endothelial Cells ,Animals ,Humans ,Kinase activity ,Molecular Biology ,Cell Proliferation ,Janus Kinases ,Tube formation ,Mice, Hairless ,Mice, Inbred BALB C ,Binding Sites ,Kinase ,Endothelial Cells ,Receptor, TIE-2 ,Vascular Endothelial Growth Factor Receptor-2 ,Xenograft Model Antitumor Assays ,Rats ,Angiogenesis inhibitor ,Vascular endothelial growth factor ,src-Family Kinases ,chemistry ,STAT protein ,Cancer research ,Janus kinase ,Signal Transduction - Abstract
Vascular endothelial growth factor receptor-2 (VEGFR-2) and Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signalling are important for tumor angiogenesis and metastasis. In this study, we identified (3-(2-(3-(morpholinomethyl)phenyl)thieno[3,2-b]pyridin-7-ylamino)phenol (LCB03-0110) as a potent angiogenesis inhibitor. LCB03-0110 inhibited VEGFR-2 and JAK/STAT3 signalling in primary cultured human endothelial cells and cancer cells. An in vitro kinase assay and molecular modelling revealed that LCB03-0110 inhibited VEGFR-2, c-SRC and TIE-2 kinase activity via preferential binding at the ATP-binding site of their kinases. LCB03-0110 successfully occupied the hydrophobic pocket of VEGFR-2, c-SRC and TIE-2. LCB03-0110 also inhibited hypoxia-induced HIF/STAT3 and EGF- or angiopoietin-induced signalling cascades. In addition, LCB03-0110 inhibited VEGF-induced proliferation, viability, migration and capillary-like tube formation. LCB03-0110 also suppressed the sprouting of endothelial cells in the rat aorta and the formation of new blood vessels in the mouse Matrigel plug assay, but also suppressed pulmonary metastasis and tumor xenograft in mice. Our results suggest that LCB03-0110 is a potential candidate small molecule for blocking angiogenesis mediated by aberrant activation of VEGFR-2 and JAK/STAT3 signalling.
- Published
- 2015
7. Promiscuous gating modifiers target the voltage sensor of K v 7.2, TRPV1, and H v 1 cation channels
- Author
-
Polina Kornilov, Sun Choi, Moshe Rehavi, Netta Roz, Bernard Attali, Karam Son, Yoonji Lee, Jin Hee Lee, Asher Peretz, and Bosmat Refaeli
- Subjects
Diclofenac ,Patch-Clamp Techniques ,Stereochemistry ,TRPV Cation Channels ,CHO Cells ,Gating ,Molecular Dynamics Simulation ,Biochemistry ,Ion Channels ,Transient receptor potential channel ,Cricetulus ,Cricetinae ,Ganglia, Spinal ,KCNQ2 Potassium Channel ,Genetics ,Animals ,Patch clamp ,Molecular Biology ,Ion channel ,Chemistry ,Chinese hamster ovary cell ,Diphenylamine ,Potassium channel ,Rats ,Electrophysiology ,Ion Channel Gating ,Biotechnology - Abstract
Some of the fascinating features of voltage-sensing domains (VSDs) in voltage-gated cation channels (VGCCs) are their modular nature and adaptability. Here we examined the VSD sensitivity of different VGCCs to 2 structurally related nontoxin gating modifiers, NH17 and NH29, which stabilize K(v)7.2 potassium channels in the closed and open states, respectively. The effects of NH17 and NH29 were examined in Chinese hamster ovary cells transfected with transient receptor potential vanilloid 1 (TRPV1) or K(v)7.2 channels, as well as in dorsal root ganglia neurons, using the whole-cell patch-clamp technique. NH17 and NH29 exert opposite effects on TRPV1 channels, operating, respectively, as an activator and a blocker of TRPV1 currents (EC50 and IC50 values ranging from 4 to 40 μM). Combined mutagenesis, electrophysiology, structural homology modeling, molecular docking, and molecular dynamics simulation indicate that both compounds target the VSDs of TRPV1 channels, which, like vanilloids, are involved in π-π stacking, H-bonding, and hydrophobic interactions. Reflecting their promiscuity, the drugs also affect the lone VSD proton channel mVSOP. Thus, the same gating modifier can promiscuously interact with different VGCCs, and subtle differences at the VSD-ligand interface will dictate whether the gating modifier stabilizes channels in either the closed or the open state.
- Published
- 2014
8. Mapping the intramolecular signal transduction of G-protein coupled receptors
- Author
-
Changbong Hyeon, Sun Choi, and Yoonji Lee
- Subjects
biology ,Chemistry ,Allosteric regulation ,Computational biology ,Bioinformatics ,Biochemistry ,Adenosine receptor ,Transmembrane protein ,Allosteric enzyme ,Structural Biology ,Rhodopsin ,biology.protein ,Signal transduction ,Receptor ,Molecular Biology ,G protein-coupled receptor - Abstract
G-protein coupled receptors (GPCRs), a major gatekeeper of extracellular signals on plasma membrane, are unarguably one of the most important therapeutic targets. Given the recent discoveries of allosteric modulations, an allosteric wiring diagram of intramolecular signal transductions would be of great use to glean the mechanism of receptor regulation. Here, by evaluating betweenness centrality (CB ) of each residue, we calculate maps of information flow in GPCRs and identify key residues for signal transductions and their pathways. Compared with preexisting approaches, the allosteric hotspots that our CB -based analysis detects for A2 A adenosine receptor (A2 A AR) and bovine rhodopsin are better correlated with biochemical data. In particular, our analysis outperforms other methods in locating the rotameric microswitches, which are generally deemed critical for mediating orthosteric signaling in class A GPCRs. For A2 A AR, the inter-residue cross-correlation map, calculated using equilibrium structural ensemble from molecular dynamics simulations, reveals that strong signals of long-range transmembrane communications exist only in the agonist-bound state. A seemingly subtle variation in structure, found in different GPCR subtypes or imparted by agonist bindings or a point mutation at an allosteric site, can lead to a drastic difference in the map of signaling pathways and protein activity. The signaling map of GPCRs provides valuable insights into allosteric modulations as well as reliable identifications of orthosteric signaling pathways.
- Published
- 2013
9. Selectivity Enhancement Arising from Interactions at the PI3K Unique Pocket
- Author
-
Kyungrok Ham, Sun Choi, Sungwoo Hong, Yujeong Jeong, Yoonji Lee, and Jung-Hyun Lee
- Subjects
Molecular model ,Pyridines ,Computational biology ,Biochemistry ,Phosphatidylinositol 3-Kinases ,Text mining ,Drug Discovery ,medicine ,Humans ,Computer Simulation ,General Pharmacology, Toxicology and Pharmaceutics ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Phosphoinositide-3 Kinase Inhibitors ,Pharmacology ,Binding Sites ,Chemistry ,business.industry ,Cyclin-Dependent Kinase 2 ,Organic Chemistry ,Cancer ,medicine.disease ,Protein Structure, Tertiary ,MCF-7 Cells ,Molecular Medicine ,Selectivity ,business - Published
- 2012
10. Link between Allosteric Signal Transduction and Functional Dynamics in a Multisubunit Enzyme: S-Adenosylhomocysteine Hydrolase
- Author
-
Yoonji Lee, Changbong Hyeon, Lak Shin Jeong, and Sun Choi
- Subjects
Protein Conformation ,Molecular Networks (q-bio.MN) ,Allosteric regulation ,Molecular Dynamics Simulation ,Ligands ,Biochemistry ,Catalysis ,Colloid and Surface Chemistry ,Protein structure ,Allosteric Regulation ,Hydrolase ,Humans ,Quantitative Biology - Molecular Networks ,Binding site ,chemistry.chemical_classification ,Binding Sites ,biology ,Chemistry ,Adenosylhomocysteinase ,Active site ,Biomolecules (q-bio.BM) ,General Chemistry ,Methylation ,Cell biology ,Protein Subunits ,Enzyme ,Quantitative Biology - Biomolecules ,FOS: Biological sciences ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
S-adenosylhomocysteine hydrolase (SAHH), a cellular enzyme that plays a key role in methylation reactions including those required for maturation of viral mRNA, is an important drug target in the discovery of antiviral agents. While targeting the active site is a straightforward strategy of enzyme inhibition, evidences of allosteric modulation of active site in many enzymes underscore the molecular origin of signal transduction. Information of co-evolving sequences in SAHH family and the key residues for functional dynamics that can be identified using native topology of the enzyme provide glimpses into how the allosteric signaling network, dispersed over the molecular structure, coordinates intra- and inter-subunit conformational dynamics. To study the link between the allosteric communication and functional dynamics of SAHHs, we performed Brownian dynamics simulations by building a coarse-grained model based on the holo and ligand-bound structures. The simulations of ligand-induced transition revealed that the signal of intra-subunit closure dynamics is transmitted to form inter-subunit contacts, which in turn invoke a precise alignment of active site, followed by the dimer-dimer rotation that compacts the whole tetrameric structure. Further analyses of SAHH dynamics associated with ligand binding provided evidence of both induced fit and population shift mechanisms, and also showed that the transition state ensemble is akin to the ligand-bound state. Besides the formation of enzyme-ligand contacts at the active site, the allosteric couplings from the residues distal to the active site is vital to the enzymatic function., 35 pages, 14 figures, 3 Tables
- Published
- 2011
11. Oxidative modifications of glyceraldehyde-3-phosphate dehydrogenase play a key role in its multiple cellular functions
- Author
-
Jin Hee Lee, Eun Joo Song, Youngmee Kim, Sun Choi, Seung Hee Yim, Kong-Joo Lee, Yoonji Lee, Na Rae Hwang, and Jaeho Jeong
- Subjects
Models, Molecular ,Biochemistry ,Structure-Activity Relationship ,Protein structure ,Nuclear Matrix-Associated Proteins ,stomatognathic system ,Catalytic Domain ,Humans ,Amino Acid Sequence ,Cysteine ,Molecular Biology ,Cells, Cultured ,Glyceraldehyde 3-phosphate dehydrogenase ,Gel electrophoresis ,Dose-Response Relationship, Drug ,biology ,Glyceraldehyde-3-Phosphate Dehydrogenases ,RNA-Binding Proteins ,RNA ,Active site ,Hydrogen Peroxide ,Cell Biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Oxidative Stress ,Cytosol ,Protein Biosynthesis ,biology.protein ,Octamer Transcription Factors ,Target protein ,Oxidation-Reduction ,Protein Processing, Post-Translational - Abstract
Knowledge of the cellular targets of ROS (reactive oxygen species) and their regulation is an essential prerequisite for understanding ROS-mediated signalling. GAPDH (glyceraldehyde-3-phosphate dehydrogenase) is known as a major target protein in oxidative stresses and becomes thiolated in its active site. However, the molecular and functional changes of oxidized GAPDH, the inactive form, have not yet been characterized. To examine the modifications of GAPDH under oxidative stress, we separated the oxidation products by two-dimensional gel electrophoresis and identified them using nanoLC-ESI-q-TOF MS/MS (nano column liquid chromatography coupled to electrospray ionization quadrupole time-of-flight tandem MS). Intracellular GAPDH subjected to oxidative stress separated into multiple acidic spots on two-dimensional gel electrophoresis and were identified as cysteine disulfide and cysteic acids on Cys152 in the active site. We identified the interacting proteins of oxidized inactive GAPDH as p54nrb (54 kDa nuclear RNA-binding protein) and PSF (polypyrimidine tract-binding protein-associated splicing factor), both of which are known to exist as heterodimers and bind to RNA and DNA. Interaction between oxidized GAPDH and p54nrb was abolished upon expression of the GAPDH active site mutant C152S. The C-terminal of p54nrb binds to GAPDH in the cytosol in a manner dependent on the dose of hydrogen peroxide. The GAPDH–p54nrb complex enhances the intrinsic topoisomerase I activation by p54nrb–PSF binding. These results suggest that GAPDH exerts other functions beyond glycolysis, and that oxidatively modified GAPDH regulates its cellular functions by changing its interacting proteins, i.e. the RNA splicing by interacting with the p54nrb–PSF complex.
- Published
- 2009
12. Capsiate, a Nonpungent Capsaicin-Like Compound, Inhibits Angiogenesis and Vascular Permeability via a Direct Inhibition of Src Kinase Activity
- Author
-
Yonghak Kim, Byung-Dong Kim, Bo Jeong Pyun, Yoonji Lee, Young Myeong Kim, Jeong Ki Min, Tae Woong Kim, Sun Choi, Tae Yoon Kim, Young Guen Kwon, and Jeong-Han Kim
- Subjects
Vascular Endothelial Growth Factor A ,Cancer Research ,Endothelium ,Angiogenesis ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Vascular permeability ,Biology ,Capillary Permeability ,Mice ,chemistry.chemical_compound ,Adenosine Triphosphate ,medicine ,Animals ,Humans ,Phosphorylation ,Protein Kinase Inhibitors ,Cells, Cultured ,Cell Proliferation ,Tube formation ,Kinase ,Autophosphorylation ,Cell biology ,Vascular endothelial growth factor ,src-Family Kinases ,medicine.anatomical_structure ,Oncology ,Biochemistry ,chemistry ,Focal Adhesion Kinase 1 ,Endothelium, Vascular ,Capsaicin ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Capsiate, a nonpungent capsaicin analogue, and its dihydroderivative dihydrocapsiate are the major capsaicinoids of the nonpungent red pepper cultivar CH-19 Sweet. In this study, we report the biological actions and underlying molecular mechanisms of capsiate on angiogenesis and vascular permeability. In vitro, capsiate and dihydrocapsiate inhibited vascular endothelial growth factor (VEGF)–induced proliferation, chemotactic motility, and capillary-like tube formation of primary cultured human endothelial cells. They also inhibited sprouting of endothelial cells in the rat aorta and formation of new blood vessels in the mouse Matrigel plug assay in response to VEGF. Moreover, both compounds blocked VEGF-induced endothelial permeability and loss of vascular endothelial (VE)–cadherin–facilitated endothelial cell-cell junctions. Importantly, capsiate suppressed VEGF-induced activation of Src kinase and phosphorylation of its downstream substrates, such as p125FAK and VE-cadherin, without affecting autophosphorylation of the VEGF receptor KDR/Flk-1. In vitro kinase assay and molecular modeling studies revealed that capsiate inhibits Src kinase activity via its preferential docking to the ATP-binding site of Src kinase. Taken together, these results suggest that capsiate could be useful for blocking pathologic angiogenesis and vascular permeability caused by VEGF. [Cancer Res 2008;68(1):227–35]
- Published
- 2008
13. Structural basis of the phosphorylation dependent complex formation of neurodegenerative disease protein Ataxin-1 and RBM17
- Author
-
Ji-Joon Song, Sun Choi, Eun Ji Kim, and Yoonji Lee
- Subjects
Models, Molecular ,Biophysics ,Ataxin 1 ,Repressor ,Peptide ,RNA-binding protein ,Nerve Tissue Proteins ,Biochemistry ,medicine ,Humans ,Spinocerebellar Ataxias ,Nuclear protein ,Surface plasmon resonance ,Phosphorylation ,Molecular Biology ,Ataxin-1 ,chemistry.chemical_classification ,biology ,Nuclear Proteins ,RNA-Binding Proteins ,Cell Biology ,Surface Plasmon Resonance ,medicine.disease ,Cell biology ,Repressor Proteins ,chemistry ,Ataxins ,Spinocerebellar ataxia ,biology.protein ,RNA Splicing Factors - Abstract
Spinocerebellar Ataxia Type1 (SCA1) is a dominantly inherited neurodegenerative disease and belongs to polyglutamine expansion disorders. The polyglutamine expansion in Ataxin-1 (ATXN1) is responsible for SCA1 pathology. ATXN1 forms at least two distinct complexes with Capicua (CIC) or RNA-binding motif protein 17 (RBM17). The wild-type ATXN1 dominantly forms a complex with CIC and the polyglutamine expanded form of ATXN1 favors to form a complex with RBM17. The phosphorylation of Ser776 in ATXN1 is critical for SCA1 pathology and serves as a binding platform for RBM17. However, the molecular basis of the phospho-specific binging of ATXN1 to RBM17 is not delineated. Here, we present the modeled structure of RBM17 bound to the phosphorylated ATXN1 peptide. The structure reveals the phosphorylation specific interaction between ATXN1 and RBM17 through a salt-bridge network. Furthermore, the modeled structure and the interactions between RBM17 and ATXN1 were validated through mutagenesis study followed by Surface Plasmon Resonance binding experiments. This work delineates the molecular basis of the interaction between RBM17 and the phosphorylated form of ATXN1, which is critical for SCA1 pathology. Furthermore, the structure of RBM17 and pATXN1 peptide might be utilized to target RBM17-ATXN1 interaction to modulate SCA1 pathogenesis.
- Published
- 2014
14. 8-Hydroxy-2-deoxyguanosine prevents plaque formation and inhibits vascular smooth muscle cell activation through Rac1 inactivation
- Author
-
Yoonji Lee, Sun Choi, Hunjoo Ha, Myung Hee Chung, Joo Young Huh, Dong Ju Son, Hwan Myung Lee, Hanjoong Jo, Jung-Hyun Lee, and Boyeon Kim
- Subjects
Male ,Models, Molecular ,rac1 GTP-Binding Protein ,Vascular smooth muscle ,Blotting, Western ,medicine.disease_cause ,Biochemistry ,Muscle, Smooth, Vascular ,Article ,Immunoenzyme Techniques ,chemistry.chemical_compound ,Mice ,Apolipoproteins E ,Cell Movement ,Superoxides ,Physiology (medical) ,medicine ,Animals ,Vasoconstrictor Agents ,Cells, Cultured ,Cell Proliferation ,chemistry.chemical_classification ,Mice, Knockout ,Reactive oxygen species ,NADPH oxidase ,biology ,Superoxide ,Angiotensin II ,Chemotaxis ,Deoxyguanosine ,Plaque, Atherosclerotic ,Cell biology ,Oxidative Stress ,chemistry ,8-Hydroxy-2'-Deoxyguanosine ,biology.protein ,Cell activation ,Reactive Oxygen Species ,Platelet-derived growth factor receptor ,Oxidative stress - Abstract
8-Hydroxy-2-deoxyguanosine (8-OHdG), a marker of oxidative stress, has been recently rediscovered to inhibit Rac1 in neutrophils and macrophages, thereby inhibiting Rac1-linked functions of these cells, including reactive oxygen species production through NADPH oxidase activation, phagocytosis, chemotaxis, and cytokine release. In vascular smooth muscle cells (VSMCs), reactive oxygen species also induce abnormal proliferation and migration leading to progression of atherosclerosis. Based upon the involvement of reactive oxygen species in phagocytic cells and VSMCs during the atherosclerotic process, we hypothesized that 8-OHdG could have antiatherosclerotic action and tested this hypothesis in an experimentally induced atherosclerosis in mice. Partially ligated ApoE knockout mice, a more physiologically relevant model of low and oscillatory flow, developed an advanced lesion in 2 weeks, and orally administered 8-OHdG significantly reduced plaque formation along with reduced superoxide formation, monocyte/macrophage infiltration, and extracellular matrix (ECM) accumulation. The effects of 8-OHdG observed in primary VSMCs were consistent with the in vivo effects of 8-OHdG and were inhibitory to angiotensin II or platelet-derived growth factor-induced production of reactive oxygen species, proliferation, migration, and ECM production. Also, angiotensin II-induced Rac1 activity in VSMCs was significantly inhibited by 8-OHdG, and transfection of constitutively active Rac1 reversed the inhibitory effect of 8-OHdG on VSMC activation. Molecular docking study showed that 8-OHdG stabilizes Rac1-GEF complex, indicating the physical contact of 8-OHdG with Rac1. These findings highly suggest that the antiatherosclerotic effect of 8-OHdG is mediated by inhibition of Rac1 activity. In conclusion, our results show a novel action of orally active 8-OHdG in suppressing atherosclerotic plaque formation in vivo and VSMC activation in vitro through inhibition of Rac1, which emphasizes a new therapeutic avenue to benefit atherosclerosis.
- Published
- 2011
15. Herbal compound farnesiferol C exerts antiangiogenic and antitumor activity and targets multiple aspects of VEGFR1 (Flt1) or VEGFR2 (Flk1) signaling cascades
- Author
-
Kwang Seok Ahn, Shi Yong Ryu, Yoonji Lee, Hyo-Jung Lee, Jae-Ho Lee, Sun Choi, Sung Hoon Kim, Hyo-Jeong Lee, Hyunsoo Bae, Kwan Hyun Kim, Eun Ok Lee, Junxuan Lu, and Kyoo Seok Ahn
- Subjects
Male ,Cancer Research ,p38 mitogen-activated protein kinases ,Angiogenesis Inhibitors ,Antineoplastic Agents ,Biology ,Pharmacology ,Focal adhesion ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Mice ,Coumarins ,Cell Line, Tumor ,Animals ,Humans ,Neoplasm Invasiveness ,Protein kinase A ,Protein kinase B ,Tube formation ,Vascular Endothelial Growth Factor Receptor-1 ,Neovascularization, Pathologic ,Kinase ,Endothelial Cells ,Vascular Endothelial Growth Factor Receptor-2 ,Rats ,Vascular endothelial growth factor ,src-Family Kinases ,Oncology ,chemistry ,Biochemistry ,cardiovascular system ,Plant Preparations ,Sesquiterpenes ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Farnesiferol C (FC) is one of the major compounds isolated from Ferula assafoetida, an Asian herbal spice used for cancer treatment as a folk remedy. Here, we examined the hypothesis that novel antiangiogenic activities of FC contribute to anticancer efficacy. In human umbilical vein endothelial cells (HUVEC), exposure to the 10 to 40 μmol/L concentration range of FC inhibited vascular endothelial growth factor (VEGF)–induced cell proliferation, migration, invasion, tube formation, and the expression of matrix metalloproteinase-2. In addition, FC inhibited the angiogenic sprouting of VEGF-treated rat aorta in an ex vivo model. Furthermore, FC inhibited the in vivo growth of mouse Lewis lung cancer allograft model by 60% (P < 0.001) at a daily i.p. dosage of 1 mg/kg body weight without any negative effect on the weight of the host mice. Immunohistochemistry staining showed decreased microvessel density (CD34) and proliferative index (Ki-67) without affecting the apoptotic (terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling) index. Mechanistically, FC decreased the binding of VEGF to VEGFR1/Flt-1, but not to VEGFR2/KDR/Flk-1. In terms of early signaling, FC exerted a rapid inhibitory action (examined within 10 minutes) on VEGF-induced autophosphorylation of VEGFR1 without affecting that of VEGFR2. Nevertheless, FC decreased the phosphorylation of most of the kinases downstream of VEGFR2: focal adhesion kinase, Src, extracellular signal-regulated kinase 1/2, p38 mitogen-activated protein kinase, and c-jun-NH2-kinase without affecting AKT. Computer simulation suggests that FC may inhibit Src or focal adhesion kinase protein activities directly through its docking to their ATP-binding sites. Taken together, the multitargeting actions of FC, particularly VEGFR1 inhibition, may make it a novel drug candidate to complement current VEGF/VEGFR2-targeting antiangiogenic modalities for cancer. Mol Cancer Ther; 9(2); 389–99
- Published
- 2010
16. Design, synthesis, and molecular modeling studies of 5'-deoxy-5'-ureidoadenosine: 5'-ureido group as multiple hydrogen bonding donor in the active site of S-adenosylhomocysteine hydrolase
- Author
-
Long Xuan Zhao, Yoonji Lee, Hea Ok Kim, Sun Choi, Dilip K. Tosh, Lak Shin Jeong, Hyung Ryong Moon, Hyun Joo Lee, Ting Wang, Shantanu Pal, and Kang Man Lee
- Subjects
Models, Molecular ,Molecular model ,Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,Drug Discovery ,Hydrolase ,Structure–activity relationship ,cardiovascular diseases ,Binding site ,Molecular Biology ,Binding Sites ,biology ,Deoxyadenosines ,Molecular Structure ,Hydrogen bond ,Chemistry ,Adenosylhomocysteinase ,Organic Chemistry ,Active site ,Hydrogen Bonding ,nervous system diseases ,Enzyme inhibitor ,Drug Design ,biology.protein ,Molecular Medicine - Abstract
5'-Deoxy-5'-ureidoadenosine was designed and synthesized as a potent inhibitor of S-adenosylhomocysteine hydrolase (SAH), in which 5'-ureido group acted as multiple hydrogen bonding donor in binding with active site residues of SAH in the molecular modeling study.
- Published
- 2007
17. Communication over the Network of Binary Switches Regulates the Activation of A2A Adenosine Receptor
- Author
-
Yoonji Lee, Changbong Hyeon, and Sun Choi
- Subjects
Agonist ,Adenosine A2 Receptor Agonists ,Receptor, Adenosine A2A ,QH301-705.5 ,medicine.drug_class ,G protein ,Molecular Networks (q-bio.MN) ,Allosteric regulation ,Molecular Dynamics Simulation ,Cellular and Molecular Neuroscience ,Molecular dynamics ,Genetics ,medicine ,Quantitative Biology - Molecular Networks ,Biology (General) ,Receptor ,Structural motif ,Molecular Biology ,Ecology, Evolution, Behavior and Systematics ,G protein-coupled receptor ,Ecology ,Chemistry ,Hydrogen Bonding ,Biomolecules (q-bio.BM) ,Adenosine receptor ,Adenosine A2 Receptor Antagonists ,Protein Subunits ,Quantitative Biology - Biomolecules ,Computational Theory and Mathematics ,Biochemistry ,FOS: Biological sciences ,Modeling and Simulation ,Biophysics ,Research Article - Abstract
Dynamics and functions of G-protein coupled receptors (GPCRs) are accurately regulated by the type of ligands that bind to the orthosteric or allosteric binding sites. To glean the structural and dynamical origin of ligand-dependent modulation of GPCR activity, we performed total $\sim$ 5 $\mu$sec molecular dynamics simulations of A$_{2A}$ adenosine receptor (A$_{2A}$AR) in its apo, antagonist-bound, and agonist-bound forms in an explicit water and membrane environment, and examined the corresponding dynamics and correlation between the 10 key structural motifs that serve as the allosteric hotspots in intramolecular signaling network. We dubbed these 10 structural motifs "binary switches" as they display molecular interactions that switch between two distinct states. By projecting the receptor dynamics on these binary switches that yield $2^{10}$ microstates, we show that (i) the receptors in apo, antagonist-bound, and agonist-bound states explore vastly different conformational space; (ii) among the three receptor states the apo state explores the broadest range of microstates; (iii) in the presence of the agonist, the active conformation is maintained through coherent couplings among the binary switches; and (iv) to be most specific, our analysis shows that W246, located deep inside the binding cleft, can serve as both an agonist sensor and actuator of ensuing intramolecular signaling for the receptor activation.Finally, our analysis of multiple trajectories generated by inserting an agonist to the apo state underscores that the transition of the receptor from inactive to active form requires the disruption of ionic-lock in the DRY motif., Comment: 28 pages, 17 figures
- Published
- 2015
18. Identification of active Plasmodium falciparum calpain to establish screening system for Pf-calpain-based drug development
- Author
-
Byoung Yul Soh, Hyun Ok Song, Sun Choi, Binna Lee, Jeong Hoon Cho, Hyun Park, Yun Young Choi, Kusuma Kaewintajuk, Jung-Hyun Lee, and Yoonji Lee
- Subjects
Models, Molecular ,Plasmodium falciparum ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,Protein Conformation ,Molecular Sequence Data ,Drug Evaluation, Preclinical ,lcsh:Infectious and parasitic diseases ,Antimalarials ,Protein structure ,Catalytic triad ,medicine ,lcsh:RC109-216 ,Homology modeling ,Amino Acid Sequence ,Calpain ,Malaria ,Monomer ,Cysteine protease ,Protease inhibitor ,Anti-malarial drug ,biology ,Sequence Homology, Amino Acid ,Research ,Active site ,biology.organism_classification ,Molecular biology ,Protease inhibitor (biology) ,Recombinant Proteins ,High-Throughput Screening Assays ,Infectious Diseases ,Biochemistry ,biology.protein ,Parasitology ,medicine.drug - Abstract
Background With the increasing resistance of malaria parasites to available drugs, there is an urgent demand to develop new anti-malarial drugs. Calpain inhibitor, ALLN, is proposed to inhibit parasite proliferation by suppressing haemoglobin degradation. This provides Plasmodium calpain as a potential target for drug development. Pf-calpain, a cysteine protease of Plasmodium falciparum, belongs to calpain-7 family, which is an atypical calpain not harboring Ca2+-binding regulatory motifs. In this present study, in order to establish the screening system for Pf-calpain specific inhibitors, the active form of Pf-calpain was first identified. Methods Recombinant Pf-calpain including catalytic subdomain IIa (rPf cal-IIa) was heterologously expressed and purified. Enzymatic activity was determined by both fluorogenic substrate assay and gelatin zymography. Molecular homology modeling was carried out to address the activation mode of Pf-calpain in the aspect of structural moiety. Results Based on the measurement of enzymatic activity and protease inhibitor assay, it was found that the active form of Pf-calpain only contains the catalytic subdomain IIa, suggesting that Pf-calpain may function as a monomeric form. The sequence prediction indicates that the catalytic subdomain IIa contains all amino acid residues necessary for catalytic triad (Cys-His-Asn) formation. Molecular modeling suggests that the Pf-calpain subdomain IIa makes an active site, holding the catalytic triad residues in their appropriate orientation for catalysis. The mutation analysis further supports that those amino acid residues are functional and have enzymatic activity. Conclusion The identified active form of Pf-calpain could be utilized to establish high-throughput screening system for Pf-calpain inhibitors. Due to its unique monomeric structural property, Pf-calpain could be served as a novel anti-malarial drug target, which has a high specificity for malaria parasite. In addition, the monomeric form of enzyme may contribute to relatively simple synthesis of selective inhibitors.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.