85 results on '"Hong Duk Youn"'
Search Results
2. Menin Enhances Androgen Receptor-Independent Proliferation and Migration of Prostate Cancer Cells
- Author
-
Taewan Kim, Kwanyoung Jeong, Eunji Kim, Kwanghyun Yoon, Jinmi Choi, Jae Hyeon Park, Jae-Hwan Kim, Hyung Sik Kim, Hong-Duk Youn, and Eun-Jung Cho
- Subjects
Male ,Receptors, Androgen ,Cell Line, Tumor ,Humans ,Prostatic Neoplasms ,Cell Biology ,General Medicine ,Molecular Biology ,Cell Proliferation ,Signal Transduction ,Transcription Factors - Abstract
The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (
- Published
- 2022
- Full Text
- View/download PDF
3. Aurkb/PP1-mediated resetting of Oct4 during the cell cycle determines the identity of embryonic stem cells
- Author
-
Jihoon Shin, Tae Wan Kim, Hyunsoo Kim, Hye Ji Kim, Min Young Suh, Sangho Lee, Han-Teo Lee, Sojung Kwak, Sang-Eun Lee, Jong-Hyuk Lee, Hyonchol Jang, Eun-Jung Cho, and Hong-Duk Youn
- Subjects
aurkb ,cell cycle of ESCs ,oct4 phosphorylation ,oct4 resetting ,PP1 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Pluripotency transcription programs by core transcription factors (CTFs) might be reset during M/G1 transition to maintain the pluripotency of embryonic stem cells (ESCs). However, little is known about how CTFs are governed during cell cycle progression. Here, we demonstrate that the regulation of Oct4 by Aurora kinase b (Aurkb)/protein phosphatase 1 (PP1) during the cell cycle is important for resetting Oct4 to pluripotency and cell cycle genes in determining the identity of ESCs. Aurkb phosphorylates Oct4(S229) during G2/M phase, leading to the dissociation of Oct4 from chromatin, whereas PP1 binds Oct4 and dephosphorylates Oct4(S229) during M/G1 transition, which resets Oct4-driven transcription for pluripotency and the cell cycle. Aurkb phosphor-mimetic and PP1 binding-deficient mutations in Oct4 alter the cell cycle, effect the loss of pluripotency in ESCs, and decrease the efficiency of somatic cell reprogramming. Our findings provide evidence that the cell cycle is linked directly to pluripotency programs in ESCs.
- Published
- 2016
- Full Text
- View/download PDF
4. Histone acylation marks respond to metabolic perturbations and enable cellular adaptation
- Author
-
Eun Jung Cho, Chanhee Jo, Eun Kyoung Kim, Jinmi Choi, Sungjoon Oh, Seokjae Park, and Hong Duk Youn
- Subjects
Cellular adaptation ,ATP citrate lyase ,Acylation ,Clinical Biochemistry ,Adaptation, Biological ,Chromatin remodelling ,Biochemistry ,Article ,Gas Chromatography-Mass Spectrometry ,Cell Line ,Epigenesis, Genetic ,Histones ,Mice ,Acetyl Coenzyme A ,Stress, Physiological ,Transcriptional regulation ,Animals ,Humans ,Metabolomics ,Coenzyme A ,Epigenetics ,Molecular Biology ,Regulation of gene expression ,biology ,Chemistry ,Acetylation ,Chromatin Assembly and Disassembly ,Chromatin ,Cell biology ,Histone ,Glucose ,Gene Expression Regulation ,biology.protein ,Metabolome ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Energy Metabolism ,Transcriptome - Abstract
Acetylation is the most studied histone acyl modification and has been recognized as a fundamental player in metabolic gene regulation, whereas other short-chain acyl modifications have only been recently identified, and little is known about their dynamics or molecular functions at the intersection of metabolism and epigenetic gene regulation. In this study, we aimed to understand the link between nonacetyl histone acyl modification, metabolic transcriptional regulation, and cellular adaptation. Using antibodies specific for butyrylated, propionylated, and crotonylated H3K23, we analyzed dynamic changes of H3K23 acylation upon various metabolic challenges. Here, we show that H3K23 modifications were highly responsive and reversibly regulated by nutrient availability. These modifications were commonly downregulated by the depletion of glucose and recovered based on glucose or fatty acid availability. Depletion of metabolic enzymes, namely, ATP citrate lyase, carnitine acetyltransferase, and acetyl-CoA synthetase, which are involved in Ac-CoA synthesis, resulted in global loss of H3K23 butyrylation, crotonylation, propionylation, and acetylation, with a profound impact on gene expression and cellular metabolic states. Our data indicate that Ac-CoA/CoA and central metabolic inputs are important for the maintenance of histone acylation. Additionally, genome-wide analysis revealed that acyl modifications are associated with gene activation. Our study shows that histone acylation acts as an immediate and reversible metabolic sensor enabling cellular adaptation to metabolic stress by reprogramming gene expression., Cellular adaptation: chromosomal protein modification acts as metabolic stress sensor Tracking the modification of a protein essential to chromosome structure could indicate the metabolic state of cells. Histone proteins provide structural support for chromosomes, and their modification influences metabolic signaling and gene expression. One possible modification adds an acyl group to the histone (acylation). Eun-Jung Cho at Sungkyunkwan University, Suwon, South Korea, and co-workers explored acylation of histone H3K23 under specific metabolic challenges, including reduced availability of glucose and metabolic enzymes. Mammalian cells rapidly alter gene expression in response to nutrient availability, enabling them to adapt under stress. The team found that H3K23 modifications were directly linked with nutrient availability and metabolic enzyme levels. H3K23 acylation specifically reprogrammed gene expression under stress conditions, suggesting that histone acylation is part of a critical sensor system that helps cells adapt to stress.
- Published
- 2020
5. The optimized core peptide derived from CABIN1 efficiently inhibits calcineurin-mediated T-cell activation
- Author
-
Sangho Lee, Han-Teo Lee, Young Ah Kim, Il-Hwan Lee, Seong-Jun Kang, Kyeongpyo Sim, Chung-Gyu Park, Kyungho Choi, and Hong-Duk Youn
- Subjects
NFATC Transcription Factors ,Calcineurin ,T-Lymphocytes ,Clinical Biochemistry ,Molecular Medicine ,Lymphocyte Activation ,Peptides ,Molecular Biology ,Biochemistry - Abstract
The C-terminal fragment of CABIN1 interacts with calcineurin and represses the transcriptional activity of the nuclear factor of activated T cells (NFAT). However, the specific sequences and mechanisms through which it binds to calcineurin are unclear. This study determined that decameric peptide (CABIN1 residues 2146–2155) is minimally required for binding to calcineurin. This peptide contains a unique “PPTP” C-terminal sequence and a “PxIxIT” N-terminal motif. Furthermore, p38MAPK phosphorylated the threonine residue of the “PPTP” sequence under physiological conditions, dramatically enhancing the peptide’s binding affinity to calcineurin. Therefore, the CABIN1 peptide inhibited the calcineurin-NFAT pathway and the activation of T cells more efficiently than the VIVIT peptide without affecting calcineurin’s phosphatase activity. The CABIN1 peptide could thus be a more potent calcineurin inhibitor and provide therapeutic opportunities for various diseases caused by the calcineurin-NFAT pathway.
- Published
- 2021
6. O-GlcNAcylation of Sox2 at threonine 258 regulates the self-renewal and early cell fate of embryonic stem cells
- Author
-
Suji Han, Hong Duk Youn, Hye Jin You, Dong Keon Kim, Eun Young Lee, Hyonchol Jang, Hansol Jang, Inyoung Hwang, Hee Yeon Kim, Jang-Seok Lee, Ji-Woong Choi, and Hyun Mu Shin
- Subjects
Threonine ,Embryonic stem cells ,Glycosylation ,Clinical Biochemistry ,Mutant ,Fluorescent Antibody Technique ,Cell fate determination ,Biology ,medicine.disease_cause ,Biochemistry ,Article ,Mice ,SOX2 ,medicine ,Animals ,Cell Lineage ,Cell Self Renewal ,Molecular Biology ,Gene ,Transcription factor ,reproductive and urinary physiology ,Alleles ,Cells, Cultured ,Gene Editing ,Mutation ,SOXB1 Transcription Factors ,Teratoma ,RNA ,Cell Differentiation ,Embryonic stem cell ,Cell biology ,Gene Expression Regulation ,embryonic structures ,Molecular Medicine ,sense organs ,biological phenomena, cell phenomena, and immunity ,Protein Processing, Post-Translational - Abstract
Sox2 is a core transcription factor in embryonic stem cells (ESCs), and O-GlcNAcylation is a type of post-translational modification of nuclear-cytoplasmic proteins. Although both factors play important roles in the maintenance and differentiation of ESCs and the serine 248 (S248) and threonine 258 (T258) residues of Sox2 are modified by O-GlcNAcylation, the function of Sox2 O-GlcNAcylation is unclear. Here, we show that O-GlcNAcylation of Sox2 at T258 regulates mouse ESC self-renewal and early cell fate. ESCs in which wild-type Sox2 was replaced with the Sox2 T258A mutant exhibited reduced self-renewal, whereas ESCs with the Sox2 S248A point mutation did not. ESCs with the Sox2 T258A mutation heterologously introduced using the CRISPR/Cas9 system, designated E14-Sox2TA/WT, also exhibited reduced self-renewal. RNA sequencing analysis under self-renewal conditions showed that upregulated expression of early differentiation genes, rather than a downregulated expression of self-renewal genes, was responsible for the reduced self-renewal of E14-Sox2TA/WT cells. There was a significant decrease in ectodermal tissue and a marked increase in cartilage tissue in E14-Sox2TA/WT-derived teratomas compared with normal E14 ESC-derived teratomas. RNA sequencing of teratomas revealed that genes related to brain development had generally downregulated expression in the E14-Sox2TA/WT-derived teratomas. Our findings using the Sox2 T258A mutant suggest that Sox2 T258 O-GlcNAc has a positive effect on ESC self-renewal and plays an important role in the proper development of ectodermal lineage cells. Overall, our study directly links O-GlcNAcylation and early cell fate decisions., Stem cell development: hold the sugar Cells that can grow into any type of cell, called embryonic stem cells (ESCs), are signaled to stay in stem cell mode (maintain stemness) by addition of a single sugar molecule to Sox2, a regulatory protein coded for by a developmental gene. Sugar modification of Sox2 was known to be involved in maintaining stemness and sometimes implicated in cancer, but the mechanism was poorly understood. When Hyonchol Jang at the National Cancer Center in South Korea and co-workers prevented sugar modification of Sox2 by changing an amino acid at the sugar-binding site, ESCs showed reduced self-renewal. Rather than repressing genes related to stemness, the modification failed to repress developmental genes, permitting cells to grow into other cell types. These results illuminate both the role of Sox2 in cancer and the importance of sugar modification in stemness.
- Published
- 2021
7. Phosphorylation of OGFOD1 by Cell Cycle-Dependent Kinase 7/9 Enhances the Transcriptional Activity of RNA Polymerase II in Breast Cancer Cells
- Author
-
Sangho Lee, Han Teo Lee, Byung Il Lee, Jae Seok Roe, Hong Duk Youn, Il Hwan Lee, Jinmi Choi, Sang Eun Lee, Sun Shin Cha, Jae Hwan Kim, Sojung Kwak, Eun Jung Cho, Bu Gyeong Kang, In Young Hwang, and Min Young Suh
- Subjects
0301 basic medicine ,Cancer Research ,RNA polymerase II ,cell cycle-dependent kinase ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cyclin-dependent kinase ,Transcription (biology) ,RNA polymerase ,Transcriptional regulation ,transcriptional regulation ,RC254-282 ,biology ,Chemistry ,Kinase ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Cell cycle ,Cell biology ,tumorigenesis ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Phosphorylation ,OGFOD1 - Abstract
Simple Summary Among the causes of accelerating cancer properties, dysregulated transcription is considerably prominent in many cancers. However, it is difficult to target transcriptional machineries due to their fundamental importance. Compared to breast cancer cell lines, we found that OGFOD1 aggravates cancers by enhancing RNA polymerase II transcriptional activity and it is improved by cell cycle-dependent kinases. Overall, we uncovered the novel mechanism for how OGFOD1 maliciously functions in breast cancers, suggesting it as a rational cancer treatment target protein. Abstract 2-oxoglutarate and iron-dependent oxygenase domain-containing protein 1 (OGFOD1) expression is upregulated in a variety of cancers and has been related to poor prognosis. However, despite this significance to cancer progression, the precise oncogenic mechanism of OGFOD1 is not understood. We demonstrated that OGFOD1 plays a role in enhancing the transcriptional activity of RNA polymerase II in breast cancer cells. OGFOD1 directly binds to the C-terminal domain of RNA polymerase II to alter phosphorylation status. The elimination of OGFOD1 resulted in decreased tumor development. Additionally, cell cycle-dependent kinase 7 and cell cycle-dependent kinase 9, critical enzymes for activating RNA polymerase II, phosphorylated serine 256 of OGFOD1, whereas a non-phosphorylated mutant OGFOD1 failed to enhance transcriptional activation and tumor growth. Consequently, OGFOD1 helps promote tumor growth by enhancing RNA polymerase II, whereas simultaneous phosphorylation of OGFOD1 by CDK enzymes is essential in stimulating RNA polymerase II-mediated transcription both in vitro and in vivo, and expression of target genes.
- Published
- 2021
8. Long non-coding RNA ChRO1 facilitates ATRX/DAXX-dependent H3.3 deposition for transcription-associated heterochromatin reorganization
- Author
-
Hong Duk Youn, Jae Hwan Kim, Han Teo Lee, Jae Hyun Yang, Keonjin Kang, Hongmin Lee, Jong-Sun Kang, Jinyoung Park, Jeung Whan Han, Byoung Ha Youn, Seon-Young Kim, Sojung Kwak, Eun Jung Cho, Hyeon Ju Jeong, and Namshik Han
- Subjects
Male ,0301 basic medicine ,X-linked Nuclear Protein ,Transcription, Genetic ,Chromosomal Proteins, Non-Histone ,Methyl-CpG-Binding Protein 2 ,Heterochromatin ,Cellular differentiation ,Cell Cycle Proteins ,Biology ,Muscle Development ,Histones ,Mice ,03 medical and health sciences ,Death-associated protein 6 ,Genetics ,Animals ,Humans ,Constitutive heterochromatin ,RNA, Small Interfering ,Muscle, Skeletal ,ATRX ,Gene Editing ,Regulation of gene expression ,Gene regulation, Chromatin and Epigenetics ,Intracellular Signaling Peptides and Proteins ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,Histone-Lysine N-Methyltransferase ,Cell biology ,Chromatin ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,NIH 3T3 Cells ,Female ,RNA, Long Noncoding ,Heterochromatin protein 1 ,CRISPR-Cas Systems ,Carrier Proteins ,Co-Repressor Proteins ,Molecular Chaperones - Abstract
Constitutive heterochromatin undergoes a dynamic clustering and spatial reorganization during myogenic differentiation. However the detailed mechanisms and its role in cell differentiation remain largely elusive. Here, we report the identification of a muscle-specific long non-coding RNA, ChRO1, involved in constitutive heterochromatin reorganization. ChRO1 is induced during terminal differentiation of myoblasts, and is specifically localized to the chromocenters in myotubes. ChRO1 is required for efficient cell differentiation, with global impacts on gene expression. It influences DNA methylation and chromatin compaction at peri/centromeric regions. Inhibition of ChRO1 leads to defects in the spatial fusion of chromocenters, and mislocalization of H4K20 trimethylation, Suv420H2, HP1, MeCP2 and cohesin. In particular, ChRO1 specifically associates with ATRX/DAXX/H3.3 complex at chromocenters to promote H3.3 incorporation and transcriptional induction of satellite repeats, which is essential for chromocenter clustering. Thus, our results unveil a mechanism involving a lncRNA that plays a role in large-scale heterochromatin reorganization and cell differentiation.
- Published
- 2018
- Full Text
- View/download PDF
9. Cyclin-dependent kinase 1 activity coordinates the chromatin associated state of Oct4 during cell cycle in embryonic stem cells
- Author
-
Eun Jung Cho, Jihoon Shin, Hyonchol Jang, Tae Wan Kim, Deog Su Hwang, Min Young Suh, In Young Hwang, Hong Duk Youn, Hye Ji Kim, Sangho Lee, and Jae Hwan Kim
- Subjects
0301 basic medicine ,G2 Phase ,Transcription, Genetic ,Biology ,environment and public health ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Protein Phosphatase 1 ,CDC2 Protein Kinase ,Genetics ,Animals ,Aurora Kinase B ,Humans ,Phosphorylation ,Mitosis ,reproductive and urinary physiology ,Cells, Cultured ,Embryonic Stem Cells ,Cyclin-dependent kinase 1 ,Chromosome decondensation ,Gene regulation, Chromatin and Epigenetics ,Cell Cycle ,Cell cycle ,Embryonic stem cell ,Chromatin ,Cell biology ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Chromosome passenger complex ,embryonic structures ,biological phenomena, cell phenomena, and immunity ,Octamer Transcription Factor-3 ,030217 neurology & neurosurgery ,Cell Division - Abstract
Cyclin-dependent kinase 1 (Cdk1) is indispensable for embryonic stem cell (ESC) maintenance and embryo development. Even though some reports have described a connection between Cdk1 and Oct4, there is no evidence that Cdk1 activity is directly linked to the ESC pluripotency transcription program. We recently reported that Aurkb/PP1-mediated Oct4 resetting is important to cell cycle maintenance and pluripotency in mouse ESCs (mESCs). In this study, we show that Cdk1 is an upstream regulator of the Oct4 phosphorylation state during cell cycle progression, and it coordinates the chromatin associated state of Oct4 for pluripotency-related gene expression within the cell cycle. Upon entry into mitosis, Aurkb in the chromosome passenger complex becomes fully activated and PP1 activity is inhibited downstream of Cdk1 activation, leading to sustaining Oct4(S229) phosphorylation and dissociation of Oct4 from chromatin during the mitotic phase. Cdk1 inhibition at the mitotic phase abnormally results in Oct4 dephosphorylation, chromosome decondensation and chromatin association of Oct4, even in replicated chromosome. Our study results suggest a molecular mechanism by which Cdk1 directly links the cell cycle to the pluripotency transcription program in mESCs.
- Published
- 2018
10. Single electron transfer by an extracellular laccase from the white-rot fungus Pleurotus ostreatus
- Author
-
Hong-Duk Youn, Kyu-Jung Kim, Jin-Soo Maeng, Young-Hoon Han, In-Beom Jeong, Gajin Jeong, Sa-Ouk Kang, and Yung Chil Hah
- Subjects
Wood-decaying fungi -- Research ,Microbial enzymes -- Research ,Electron transport -- Analysis ,Biological sciences - Abstract
EPR and absorption spectroscopy help characterize an extracellular laccase I from the white-rot fungus Pleurotus ostreatus. Two ionization groups function as the ligand of copper metal in the active site of the laccase I. Analysis of the action of laccase I on 3,5-dimethoxy-5-hydroxyacetophenone by EPR spectroscopy reveals the involvement of the phenoxy radical as an intermediate in the single electron transfer catalyzed by laccase I.
- Published
- 1995
11. Phosphorylation of OCT4 Serine 236 Inhibits Germ Cell Tumor Growth by Inducing Differentiation
- Author
-
Hyonchol Jang, Han Seong Kim, Kyeong Man Hong, Seon Hyeong Lee, Hong Duk Youn, Soo Youl Kim, Sang Won Kang, Bomin Song, Seungjin Lee, Dong Keon Kim, Jong Kwang Kim, Hansol Jang, Seung Hyun Bae, Byung Il Lee, Suji Han, and Hee Yeon Kim
- Subjects
0301 basic medicine ,Cancer Research ,cells ,Cell ,Endogeny ,Oct4 ,cancer differentiation ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,medicine ,OCT4 serine 236 ,Transcription factor ,reproductive and urinary physiology ,phosphorylation ,Chemistry ,fungi ,germ cell tumor ,Protein phosphatase 1 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,embryonic structures ,Phosphorylation ,biological phenomena, cell phenomena, and immunity ,Germ cell ,OCT4 inhibitor - Abstract
Simple Summary Octamer-binding transcription factor 4 (OCT4) plays an important role in early embryonic development, but is rarely expressed in adults. However, in many cancer cells, this gene is re-expressed, making the cancer malignant. This present study revealed that inhibiting OCT4 transcriptional activity induces cancer cell differentiation and growth retardation. Specifically, when the phosphorylation of OCT4 serine 236 increases by interfering with the binding of protein phosphatase 1 (PP1) to OCT4, OCT4 loses its transcriptional activity and cancer cells differentiate. Therefore, this study presents the basis for the development of protein-protein interaction inhibitors that inhibit the binding of OCT4 and PP1 for cancer treatment. Abstract Octamer-binding transcription factor 4 (Oct4) plays an important role in maintaining pluripotency in embryonic stem cells and is closely related to the malignancies of various cancers. Although posttranslational modifications of Oct4 have been widely studied, most of these have not yet been fully characterized, especially in cancer. In this study, we investigated the role of phosphorylation of serine 236 of OCT4 [OCT4 (S236)] in human germ cell tumors (GCTs). OCT4 was phosphorylated at S236 in a cell cycle-dependent manner in a patient sample and GCT cell lines. The substitution of endogenous OCT4 by a mimic of phosphorylated OCT4 with a serine-to-aspartate mutation at S236 (S236D) resulted in tumor cell differentiation, growth retardation, and inhibition of tumor sphere formation. GCT cells expressing OCT4 S236D instead of endogenous OCT4 were similar to cells with OCT4 depletion at the mRNA transcript level as well as in the phenotype. OCT4 S236D also induced tumor cell differentiation and growth retardation in mouse xenograft experiments. Inhibition of protein phosphatase 1 by chemicals or short hairpin RNAs increased phosphorylation at OCT4 (S236) and resulted in the differentiation of GCTs. These results reveal the role of OCT4 (S236) phosphorylation in GCTs and suggest a new strategy for suppressing OCT4 in cancer.
- Published
- 2020
- Full Text
- View/download PDF
12. Abundance of C-terminal binding protein isoform is a prerequisite for exit from pluripotency in mouse embryonic stem cells
- Author
-
Han Teo Lee, Eun Jung Cho, Hyonchol Jang, Seong-Tae Kim, Min Young Suh, Jihoon Shin, Jae Hwan Kim, Hong Duk Youn, Tae Wan Kim, and Hye Ji Kim
- Subjects
0301 basic medicine ,Gene isoform ,Gene knockdown ,Binding protein ,Cell fate determination ,Biology ,Biochemistry ,Embryonic stem cell ,CTBP2 ,Cell biology ,03 medical and health sciences ,CTBP1 ,030104 developmental biology ,0302 clinical medicine ,Genetics ,Epigenetics ,Molecular Biology ,030217 neurology & neurosurgery ,Biotechnology - Abstract
Unlike lower organisms, mammals have 2 C-terminal binding protein (Ctbp) isoforms, Ctbp1 and Ctbp2. Ctbp2 is revealed as a key factor involved in determining cell fate decisions by regulating the epigenetic state in active embryonic stem cell (ESC) genes. However, the molecular mechanism underlying how Ctbp1 and Ctbp2 have different roles remains elusive. Here we demonstrate that Ctbp isoform abundance is important for mouse embryonic ESCs (mESCs) to exit from pluripotency. Temporal expression patterns of Ctbp isoforms were quite different; Ctbp2 is more highly expressed in mESCs and decreases during differentiation, while Ctbp1 is constantly expressed at a lower level. Ctbp2 knockdown, but not Ctbp1 knockdown, in mESCs resulted in impaired exit from pluripotency. Interestingly, Ctbp1 and Ctbp2 overexpression in Ctbp2-knockdown mESCs leads to exiting from pluripotency in a manner similar to that of wild-type mESCs. Quantification of Ctbp1 and Ctbp2 revealed that differentiation ability correlates with abundance of Ctbp isoform in undifferentiated mESCs, suggesting that a sufficient amount of Ctbp isoform is a prerequisite for exiting from pluripotency. The results support the contention that 2 redundant Ctbp isoforms regulate elaborate differentiation via temporally distinctive regulatory patterns in mESCs.-Suh, M. Y., Kim, T. W., Lee, H.-T., Shin, J., Kim, J.-H., Jang, H., Kim, H. J., Kim, S.-T., Cho, E.-J., Youn, H.-D. Abundance of C-terminal binding protein isoform is a prerequisite for exit from pluripotency in mouse embryonic stem cells.
- Published
- 2018
13. AKT phosphorylates H3-threonine 45 to facilitate termination of gene transcription in response to DNA damage
- Author
-
Hyonchol Jang, Hong Duk Youn, Sojung Kwak, Byung Hee Kang, J. K. Han, Eun Jung Cho, Tae Wan Kim, Jinmi Choi, and Jong Hyuk Lee
- Subjects
Threonine ,General transcription factor ,Termination factor ,Gene regulation, Chromatin and Epigenetics ,RNA polymerase II ,Biology ,Molecular biology ,Cell Line ,Histones ,Terminator (genetics) ,Transcription (biology) ,Transcription Termination, Genetic ,MCF-7 Cells ,Genetics ,biology.protein ,Humans ,Phosphorylation ,Transcription Initiation Site ,Transcription factor II D ,Proto-Oncogene Proteins c-akt ,Transcription factor II B ,RNA polymerase II holoenzyme ,DNA Damage ,HeLa Cells - Abstract
Post-translational modifications of core histones affect various cellular processes, primarily through transcription. However, their relationship with the termination of transcription has remained largely unknown. In this study, we show that DNA damage-activated AKT phosphorylates threonine 45 of core histone H3 (H3-T45). By genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) analysis, H3-T45 phosphorylation was distributed throughout DNA damage-responsive gene loci, particularly immediately after the transcription termination site. H3-T45 phosphorylation pattern showed close-resemblance to that of RNA polymerase II C-terminal domain (CTD) serine 2 phosphorylation, which establishes the transcription termination signal. AKT1 was more effective than AKT2 in phosphorylating H3-T45. Blocking H3-T45 phosphorylation by inhibiting AKT or through amino acid substitution limited RNA decay downstream of mRNA cleavage sites and decreased RNA polymerase II release from chromatin. Our findings suggest that AKT-mediated phosphorylation of H3-T45 regulates the processing of the 3′ end of DNA damage-activated genes to facilitate transcriptional termination.
- Published
- 2015
- Full Text
- View/download PDF
14. OGFOD1 is required for breast cancer cell proliferation and is associated with poor prognosis in breast cancer
- Author
-
Sohyun Chun, Jae Hwan Kim, Eun Jung Cho, Hong Duk Youn, Jae Seok Roe, Jong Hyuk Lee, Sojung Kwak, Hyunsoo Kim, Byung Hee Kang, Soon Min Lee, Tae Wan Kim, and Woo Ho Kim
- Subjects
Gerontology ,Time Factors ,Translational termination ,Breast Neoplasms ,Cell Cycle Proteins ,Kaplan-Meier Estimate ,Transfection ,Breast cancer ,breast cancer ,Databases, Genetic ,medicine ,Humans ,RNA, Messenger ,Nuclear protein ,Transcription factor ,In Situ Hybridization ,Cell Proliferation ,Gene knockdown ,Cell growth ,business.industry ,Computational Biology ,Nuclear Proteins ,G2/M phase ,Cell cycle ,medicine.disease ,Chromatin Assembly and Disassembly ,Prognosis ,G1 Phase Cell Cycle Checkpoints ,Immunohistochemistry ,G2 Phase Cell Cycle Checkpoints ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,Oncology ,Cytoplasm ,Gene Knockdown Techniques ,Cancer research ,MCF-7 Cells ,OGFOD1 ,cell cycle ,Female ,RNA Interference ,business ,Carrier Proteins ,Research Paper ,HeLa Cells ,Signal Transduction ,Transcription Factors - Abstract
2-oxogluatrate and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) was recently revealed to be a proline hydroxylase of RPS23 for translational termination. However, OGFOD1 is nuclear, whereas translational termination occurs in the cytoplasm, raising the possibility of another function of OGFOD1 in the nucleus. In this study, we demonstrate that OGFOD1 is involved in cell cycle regulation. OGFOD1 knockdown in MDA-MB-231 breast cancer cells significantly impeded cell proliferation and resulted in the accumulation of G1 and G2/M cells by decreasing the mRNA levels of G1/S transition- and G2/M-related transcription factors and their target genes. We also confirmed that OGFOD1 is highly expressed in breast cancer tissues by bioinformatic analysis and immunohistochemistry. Thus, we propose that OGFOD1 is required for breast cancer cell proliferation and is associated with poor prognosis in breast cancer.
- Published
- 2015
15. A KAP1 phosphorylation switch controls MyoD function during skeletal muscle differentiation
- Author
-
Suk Min Jang, Evarist Planet, F. Jeffrey Dilworth, Didier Trono, Hervé Faralli, Hong Duk Youn, Jinmi Choi, Marco Cassano, Gurjeev Sohi, Kulwant Singh, and Soji Sebastian
- Subjects
TRIM28 ,G9a ,animal structures ,Cellular differentiation ,Tripartite Motif-Containing Protein 28 ,Biology ,Muscle Development ,MyoD ,Cell Line ,Myoblasts ,Mice ,03 medical and health sciences ,0302 clinical medicine ,MyoD Protein ,Genetics ,Animals ,Myocyte ,Phosphorylation ,Muscle, Skeletal ,Myogenin ,030304 developmental biology ,0303 health sciences ,PITX2 ,epigenetics ,MEF2 Transcription Factors ,Myogenesis ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Cell Differentiation ,musculoskeletal system ,Repressor Proteins ,enzymes and coenzymes (carbohydrates) ,KAP1 ,Cancer research ,myogenesis ,MSK1 phosphorylation ,tissues ,030217 neurology & neurosurgery ,Research Paper ,Signal Transduction ,Developmental Biology - Abstract
The transcriptional activator MyoD serves as a master controller of myogenesis. Often in partnership with Mef2 (myocyte enhancer factor 2), MyoD binds to the promoters of hundreds of muscle genes in proliferating myoblasts yet activates these targets only upon receiving cues that launch differentiation. What regulates this off/on switch of MyoD function has been incompletely understood, although it is known to reflect the action of chromatin modifiers. Here, we identify KAP1 (KRAB [Krüppel-like associated box]-associated protein 1)/TRIM28 (tripartite motif protein 28) as a key regulator of MyoD function. In myoblasts, KAP1 is present with MyoD and Mef2 at many muscle genes, where it acts as a scaffold to recruit not only coactivators such as p300 and LSD1 but also corepressors such as G9a and HDAC1 (histone deacetylase 1), with promoter silencing as the net outcome. Upon differentiation, MSK1-mediated phosphorylation of KAP1 releases the corepressors from the scaffold, unleashing transcriptional activation by MyoD/Mef2 and their positive cofactors. Thus, our results reveal KAP1 as a previously unappreciated interpreter of cell signaling, which modulates the ability of MyoD to drive myogenesis.
- Published
- 2015
16. Zinc finger proteins orchestrate active gene silencing during embryonic stem cell differentiation
- Author
-
Jang Seok Lee, Jong Hyuk Lee, Jihoon Shin, Hong Duk Youn, Sojung Kwak, In Young Hwang, Han Teo Lee, Eun Jung Cho, Jae Hwan Kim, Byung Hee Kang, and Tae Wan Kim
- Subjects
0301 basic medicine ,Transcription, Genetic ,Nerve Tissue Proteins ,Biology ,Chromatin remodeling ,03 medical and health sciences ,Mice ,Genetics ,Gene silencing ,Animals ,Humans ,Epigenetics ,Gene Silencing ,Transcription factor ,reproductive and urinary physiology ,Cells, Cultured ,Embryonic Stem Cells ,Zinc finger ,urogenital system ,Gene regulation, Chromatin and Epigenetics ,Cell Differentiation ,Mi-2/NuRD complex ,Cell biology ,Chromatin ,Alcohol Oxidoreductases ,030104 developmental biology ,embryonic structures ,biology.protein ,Trans-Activators ,biological phenomena, cell phenomena, and immunity ,PRC2 ,Co-Repressor Proteins - Abstract
Transcription factors and chromatin remodeling proteins control the transcriptional variability for ESC lineage commitment. During ESC differentiation, chromatin modifiers are recruited to the regulatory regions by transcription factors, thereby activating the lineage-specific genes or silencing the transcription of active ESC genes. However, the underlying mechanisms that link transcription factors to exit from pluripotency are yet to be identified. In this study, we show that the Ctbp2-interacting zinc finger proteins, Zfp217 and Zfp516, function as linkers for the chromatin regulators during ESC differentiation. CRISPR-Cas9-mediated knock-outs of both Zfp217 and Zfp516 in ESCs prevent the exit from pluripotency. Both zinc finger proteins regulate the Ctbp2-mediated recruitment of the NuRD complex and polycomb repressive complex 2 (PRC2) to active ESC genes, subsequently switching the H3K27ac to H3K27me3 during ESC differentiation for active gene silencing. We therefore suggest that some zinc finger proteins orchestrate to control the concise epigenetic states on active ESC genes during differentiation, resulting in natural lineage commitment.
- Published
- 2017
17. PHF2 histone demethylase acts as a tumor suppressor in association with p53 in cancer
- Author
-
H. S. Sung, Hong Duk Youn, Yang Suk Chun, Chung-Hyun Cho, Sang Jeong Kim, Yong Joon Choi, Jong Wan Park, Kil-Song Lee, Hyo-Suk Lee, Hyun Woo Shin, S. H. Li, Woo Ho Kim, H. J. Chung, and Tae-You Kim
- Subjects
Cancer Research ,Colorectal cancer ,Mice, Nude ,Biology ,Mice ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Humans ,Genes, Tumor Suppressor ,Epigenetics ,RNA, Small Interfering ,Stomach cancer ,Molecular Biology ,Homeodomain Proteins ,Regulation of gene expression ,Cell Death ,Hep G2 Cells ,HCT116 Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Chromatin ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,Cancer cell ,Chromosomal region ,MCF-7 Cells ,biology.protein ,Cancer research ,Demethylase ,Tumor Suppressor Protein p53 - Abstract
Plant homeodomain finger 2 (PHF2) has a role in epigenetic regulation of gene expression by demethylating H3K9-Me2. Several genome-wide studies have demonstrated that the chromosomal region including the PHF2 gene is often deleted in some cancers including colorectal cancer, and this finding encouraged us to investigate the tumor suppressive role of PHF2. As p53 is a critical tumor suppressor in colon cancer, we tested the possibility that PHF2 is an epigenetic regulator of p53. PHF2 was associated with p53, and thereby, promoted p53-driven gene expression in cancer cells under genotoxic stress. PHF2 converted the chromatin that is favorable for transcription by demethylating the repressive H3K9-Me2 mark. In an HCT116 xenograft model, PHF2 was found to be required for the anticancer effects of oxaliplatin and doxorubicin. In PHF2-deficient xenografts, p53 expression was profoundly induced by both drugs, but its downstream product p21 was not, suggesting that p53 cannot be activated in the absence of PHF2. To find clinical evidence about the role of PHF2, we analyzed the expressions of PHF2, p53 and p21 in human colon cancer tissues and adjacent normal tissues from patients. PHF2 was downregulated in cancer tissues and PHF2 correlated with p21 in cancers expressing functional p53. Colon and stomach cancer tissue arrays showed a positive correlation between PHF2 and p21 expressions. Informatics analyses using the Oncomine database also supported our notion that PHF2 is downregulated in colon and stomach cancers. On the basis of these findings, we propose that PHF2 acts as a tumor suppressor in association with p53 in cancer development and ensures p53-mediated cell death in response to chemotherapy.
- Published
- 2014
- Full Text
- View/download PDF
18. Modulation of lysine methylation in myocyte enhancer factor 2 during skeletal muscle cell differentiation
- Author
-
Eun Jung Cho, Hyonchol Jang, Seong-Tae Kim, Jong Hyuk Lee, Jinmi Choi, Hyunsoo Kim, and Hong Duk Youn
- Subjects
Mef2 ,animal structures ,Transcription, Genetic ,Myoblasts, Skeletal ,Gene Regulation, Chromatin and Epigenetics ,Biology ,Methylation ,Cell Line ,Mice ,Skeletal muscle cell differentiation ,Epigenetics of physical exercise ,Histone methylation ,Genetics ,Animals ,Humans ,Histone Demethylases ,MEF2 Transcription Factors ,Lysine ,EZH2 ,Pioneer factor ,Cell Differentiation ,Oxidoreductases, N-Demethylating ,Histone-Lysine N-Methyltransferase ,musculoskeletal system ,Chromatin ,HEK293 Cells ,Biochemistry ,Histone methyltransferase ,Protein Processing, Post-Translational - Abstract
Myocyte enhancer factor 2 (MEF2) is a family of transcription factors that regulates many processes, including muscle differentiation. Due to its many target genes, MEF2D requires tight regulation of transcription activity over time and by location. Epigenetic modifiers have been suggested to regulate MEF2-dependent transcription via modifications to histones and MEF2. However, the modulation of MEF2 activity by lysine methylation, an important posttranslational modification that alters the activities of transcription factors, has not been studied. We report the reversible lysine methylation of MEF2D by G9a and LSD1 as a regulatory mechanism of MEF2D activity and skeletal muscle differentiation. G9a methylates lysine-267 of MEF2D and represses its transcriptional activity, but LSD1 counteracts it. This residue is highly conserved between MEF2 members in mammals. During myogenic differentiation of C2C12 mouse skeletal muscle cells, the methylation of MEF2D by G9a decreased, on which MEF2D-dependent myogenic genes were upregulated. We have also identified lysine-267 as a methylation/demethylation site and demonstrate that the lysine methylation state of MEF2D regulates its transcriptional activity and skeletal muscle cell differentiation.
- Published
- 2013
- Full Text
- View/download PDF
19. Dissecting the roles of the histone chaperones reveals the evolutionary conserved mechanism of transcription-coupled deposition of H3.3
- Author
-
Jae Hyun Yang, Yunkyoung Song, Hye Jin Kim, Jeung Whan Han, Hong Duk Youn, Eun Jung Cho, and Ja Hwan Seol
- Subjects
Transcription, Genetic ,Euchromatin ,Heterochromatin ,Cell Cycle Proteins ,Gene Regulation, Chromatin and Epigenetics ,Conserved sequence ,Fungal Proteins ,Histones ,Histone H3 ,Transcription (biology) ,Yeasts ,HIR complex ,Genetics ,Humans ,Histone Chaperones ,Amino Acid Sequence ,Conserved Sequence ,biology ,Biological Evolution ,Protein Structure, Tertiary ,Chromatin ,Histone ,Mutation ,biology.protein ,Transcription Factors - Abstract
The mammalian genome encodes multiple variants of histone H3 including H3.1/H3.2 and H3.3. In contrast to H3.1/H3.2, H3.3 is enriched in the actively transcribed euchromatin and the telomeric heterochromatins. However, the mechanism for H3.3 to incorporate into the different domains of chromatin is not known. Here, taking the advantage of well-defined transcription analysis system of yeast, we attempted to understand the molecular mechanism of selective deposition of human H3.3 into actively transcribed genes. We show that there are systemic H3 substrate-selection mechanisms operating even in yeasts, which encode a single type of H3. Yeast HIR complex mediated H3-specific recognition specificity for deposition of H3.3 in the transcribed genes. A critical component of this process was the H3 A-IG code composed of amino acids 87, 89 and 90. The preference toward H3.3 was completely lost when HIR subunits were absent and partially suppressed by human HIRA. Asf1 allows the influx of H3, regardless of H3 type. We propose that H3.3 is introduced into the active euchromatin by targeting the recycling pathway that is mediated by HIRA (or HIR), and this H3-selection mechanism is highly conserved through the evolution. These results also uncover an unexpected role of RI chaperones in evolution of variant H3s.
- Published
- 2013
- Full Text
- View/download PDF
20. Phosphorylation and ubiquitination-dependent degradation of CABIN1 releases p53 for transactivation upon genotoxic stress
- Author
-
Hong Duk Youn, Jae Seok Roe, Soo Youn Choi, Seong-Tae Kim, Eun Jung Cho, and Hyonchol Jang
- Subjects
Transcriptional Activation ,Ubiquitin-Protein Ligases ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Genotoxic Stress ,Gene Regulation, Chromatin and Epigenetics ,Protein Serine-Threonine Kinases ,Ubiquitin ,Stress, Physiological ,Genetics ,Humans ,Phosphorylation ,Adaptor Proteins, Signal Transducing ,Protein-Serine-Threonine Kinases ,biology ,Kinase ,Tumor Suppressor Proteins ,Ubiquitination ,Chromatin ,Cell biology ,DNA-Binding Proteins ,Checkpoint Kinase 2 ,Biochemistry ,Ubiquitin ligase complex ,Proteolysis ,biology.protein ,Tumor Suppressor Protein p53 ,DNA Damage ,Mutagens - Abstract
CABIN1 acts as a negative regulator of p53 by keeping p53 in an inactive state on chromatin. Genotoxic stress causes rapid dissociation of CABIN1 and activation of p53. However, its molecular mechanism is still unknown. Here, we reveal the phosphorylation- and ubiquitination-dependent degradation of CABIN1 upon DNA damage, releasing p53 for transcriptional activation. The DNA-damage-signaling kinases, ATM and CHK2, phosphorylate CABIN1 and increase the degradation of CABIN1 protein. Knockdown or overexpression of these kinases influences the stability of CABIN1 protein showing that their activity is critical for degradation of CABIN1. Additionally, CABIN1 was found to undergo ubiquitin-dependent proteasomal degradation mediated by the CRL4DDB2 ubiquitin ligase complex. Both phosphorylation and ubiquitination of CABIN1 appear to be relevant for controlling the level of CABIN1 protein upon genotoxic stress.
- Published
- 2013
- Full Text
- View/download PDF
21. Psat1-Dependent Fluctuations in α-Ketoglutarate Affect the Timing of ESC Differentiation
- Author
-
Young A. Kim, Yoon Kyung Jeon, Hong Duk Youn, Hyonchol Jang, Sojung Kwak, Hyunsoo Kim, Sunghyouk Park, Jae Hwan Kim, Eun Jung Cho, Xing Jin, In Young Hwang, Doo Hyun Chung, Sangho Lee, and Sang Eun Lee
- Subjects
0301 basic medicine ,Homeobox protein NANOG ,Time Factors ,Physiology ,Intracellular Space ,Biology ,Histones ,03 medical and health sciences ,Mice ,SOX2 ,Histone methylation ,Animals ,Phosphoserine Aminotransferase ,Epigenetics ,Molecular Biology ,Transaminases ,Gene knockdown ,Cell Differentiation ,Mouse Embryonic Stem Cells ,Cell Biology ,DNA Methylation ,Molecular biology ,Embryonic stem cell ,030104 developmental biology ,embryonic structures ,Ketoglutaric Acids ,Intracellular ,Transcription Factors - Abstract
Summary Embryonic stem cells (ESCs) undergo coordinated epigenetic and metabolic changes to differentiate properly. However, the precise mechanisms by which these alterations are fine-tuned in the early stages of differentiation have not been identified. In this study, we demonstrate that phosphoserine aminotransferase 1 (Psat1), an Oct4/Sox2/Nanog (OSN) target protein, regulates changes in α-ketoglutarate (α-KG), determining the fate of mouse ESCs (mESCs). Maintaining Psat1 levels was essential for mESC self-renewal and pluripotency. Moderate knockdown (KD) of Psat1 in mESCs lowered DNA 5′-hydroxymethylcytosine (5′-hmC) and increased histone methylation levels by downregulating permissive amounts of α-KG, ultimately accelerating differentiation. We found that intracellular α-KG declined transiently during differentiation and that its dysregulation by treatment with dimethyl-α-KG impeded differentiation. Further, by in vivo teratoma formation assay, pluripotency of Psat1 KD mESCs was impaired, especially into the ectodermal lineage. Thus, we have established how Psat1 is regulated in maintaining intracellular α-KG levels and determining the fate of mESCs.
- Published
- 2016
22. Aurkb/PP1-mediated resetting of Oct4 during the cell cycle determines the identity of embryonic stem cells
- Author
-
Hyonchol Jang, Jong Hyuk Lee, Hye Ji Kim, Sangho Lee, Jihoon Shin, Sang Eun Lee, Hong Duk Youn, Sojung Kwak, Eun Jung Cho, Hyunsoo Kim, Min Young Suh, Tae Wan Kim, and Han Teo Lee
- Subjects
0301 basic medicine ,oct4 resetting ,Mouse ,QH301-705.5 ,Cellular differentiation ,Rex1 ,cells ,Science ,Embryoid body ,Biology ,Biochemistry ,General Biochemistry, Genetics and Molecular Biology ,cell cycle of ESCs ,03 medical and health sciences ,oct4 phosphorylation ,Mice ,Protein Phosphatase 1 ,aurkb ,Animals ,Aurora Kinase B ,Biology (General) ,Phosphorylation ,Cell potency ,reproductive and urinary physiology ,Embryonic Stem Cells ,General Immunology and Microbiology ,General Neuroscience ,fungi ,Cell Cycle ,General Medicine ,Cell Biology ,Cell cycle ,PP1 ,Embryonic stem cell ,Cell Cycle Gene ,Cell biology ,030104 developmental biology ,embryonic structures ,Medicine ,biological phenomena, cell phenomena, and immunity ,Reprogramming ,Octamer Transcription Factor-3 ,Protein Processing, Post-Translational ,Research Article - Abstract
Pluripotency transcription programs by core transcription factors (CTFs) might be reset during M/G1 transition to maintain the pluripotency of embryonic stem cells (ESCs). However, little is known about how CTFs are governed during cell cycle progression. Here, we demonstrate that the regulation of Oct4 by Aurora kinase b (Aurkb)/protein phosphatase 1 (PP1) during the cell cycle is important for resetting Oct4 to pluripotency and cell cycle genes in determining the identity of ESCs. Aurkb phosphorylates Oct4(S229) during G2/M phase, leading to the dissociation of Oct4 from chromatin, whereas PP1 binds Oct4 and dephosphorylates Oct4(S229) during M/G1 transition, which resets Oct4-driven transcription for pluripotency and the cell cycle. Aurkb phosphor-mimetic and PP1 binding-deficient mutations in Oct4 alter the cell cycle, effect the loss of pluripotency in ESCs, and decrease the efficiency of somatic cell reprogramming. Our findings provide evidence that the cell cycle is linked directly to pluripotency programs in ESCs. DOI: http://dx.doi.org/10.7554/eLife.10877.001, eLife digest Embryonic stem cells can give rise to any type of cell in the body – an ability known as pluripotency. These cells rapidly divide and self-renew until they are exposed to signals that cause them to mature into a particular specialized cell type. As cells prepare to divide, they transition through a series of phases known as the cell cycle. In embryonic stem cells, these phases are often shorter than in other cell types. This altered timing is thought to be important for maintaining the pluripotency of the stem cells. Proteins called core transcription factors also help stem cells to remain pluripotent. Evidence suggests that the activity of some of these proteins affects the timing of the different cell cycle phases. However, it is not clear exactly how they do so or how the activity of the transcription factors is controlled. A core transcription factor called Oct4 is thought to be a “master regulator” of pluripotency that controls the activity of many of the other core transcription factors. Shin, Kim, Kim, Kim et al. have now studied the activity of Oct4 around the point of cell division. This revealed that a protein called aurora kinase B modifies Oct4 by adding a phosphate group to it just before a cell divides. This modification causes Oct4 to detach from chromatin, the protein structure in which DNA is packaged inside cells. Following cell division, another protein called PP1 removes the phosphate group from Oct4. This “resets” the pluripotency of the stem cell, allowing it to continue to self-renew. Cells that contain only mutant forms of Oct4 that cannot bind to aurora kinase B or PP1 lose their pluripotency. The mutant Oct4 proteins also alter the cell cycle of the stem cells. Overall, Shin et al.’s findings suggest that Oct4 regulates the cell cycle of embryonic stem cells as well as their pluripotency. How Oct4 activity affects the specialization of the stem cells into mature cell types remains to be investigated in future studies. DOI: http://dx.doi.org/10.7554/eLife.10877.002
- Published
- 2016
23. O-GlcNAc Regulates Pluripotency and Reprogramming by Directly Acting on Core Components of the Pluripotency Network
- Author
-
Tae-Wook Kang, Tae Wan Kim, Yoo Wook Kwon, Soo Youn Choi, Hong Duk Youn, Sungho Yoon, Eun Jung Cho, Hyonchol Jang, and Seon-Young Kim
- Subjects
Pluripotent Stem Cells ,Glycosylation ,Transcription, Genetic ,Somatic cell ,Molecular Sequence Data ,Regulator ,Biology ,Models, Biological ,Acetylglucosamine ,Mice ,SOX2 ,Genetics ,Animals ,Humans ,Gene Regulatory Networks ,Amino Acid Sequence ,Induced pluripotent stem cell ,reproductive and urinary physiology ,Embryonic Stem Cells ,Cell Proliferation ,Regulation of gene expression ,SOXB1 Transcription Factors ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,Cellular Reprogramming ,Embryonic stem cell ,Cell biology ,Mice, Inbred C57BL ,KLF2 ,embryonic structures ,Mutation ,Molecular Medicine ,biological phenomena, cell phenomena, and immunity ,Reprogramming ,Octamer Transcription Factor-3 - Abstract
SummaryO-linked-N-acetylglucosamine (O-GlcNAc) has emerged as a critical regulator of diverse cellular processes, but its role in embryonic stem cells (ESCs) and pluripotency has not been investigated. Here we show that O-GlcNAcylation directly regulates core components of the pluripotency network. Blocking O-GlcNAcylation disrupts ESC self-renewal and reprogramming of somatic cells to induced pluripotent stem cells. The core reprogramming factors Oct4 and Sox2 are O-GlcNAcylated in ESCs, but the O-GlcNAc modification is rapidly removed upon differentiation. O-GlcNAc modification of threonine 228 in Oct4 regulates Oct4 transcriptional activity and is important for inducing many pluripotency-related genes, including Klf2, Klf5, Nr5a2, Tbx3, and Tcl1. A T228A point mutation that eliminates this O-GlcNAc modification reduces the capacity of Oct4 to maintain ESC self-renewal and reprogram somatic cells. Overall, our study makes a direct connection between O-GlcNAcylation of key regulatory transcription factors and the activity of the pluripotency network.
- Published
- 2012
- Full Text
- View/download PDF
24. Phosphorylation of von Hippel-Lindau protein by checkpoint kinase 2 regulates p53 transactivation
- Author
-
Hong Duk Youn, Jae Seok Roe, In Young Hwang, Seong-Tae Kim, Eun Jung Cho, Hwa Ryeon Kim, and Nam-Chul Ha
- Subjects
Transcriptional Activation ,DNA damage ,Apoptosis ,Protein Serine-Threonine Kinases ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,Lysine Acetyltransferase 5 ,Transactivation ,Serine ,medicine ,Humans ,Phosphorylation ,Molecular Biology ,Checkpoint Kinase 2 ,Transcription factor ,Histone Acetyltransferases ,Cell Biology ,HCT116 Cells ,female genital diseases and pregnancy complications ,Chromatin ,Von Hippel-Lindau Tumor Suppressor Protein ,Cancer research ,Tumor Suppressor Protein p53 ,Carcinogenesis ,E1A-Associated p300 Protein ,DNA Damage ,Developmental Biology - Abstract
von-Hippel Lindau protein (pVHL) suppresses tumorigenesis in the kidney, in part through regulation of hypoxia-inducible factor alpha (HIF alpha). However, HIF has been proposed to be necessary but insufficient for renal tumorigenesis. p53 was implicated as a transcription factor that is regulated by pVHL, but the molecular mechanism by which pVHL regulates p53 on DNA damage is unknown. We demonstrated that checkpoint kinase-2 (Chk2) binds to the beta-domain of pVHL and phosphorylates Ser 111 on DNA damage. Notably, this modification enhances pVHL-mediated transactivation of p53 by recruiting p300 and Tip60 to the chromatin of p53 target gene. Further, the naturally occurring pVHL mutants pVHL-S111R and pVHL-S111C showed diminished binding to coactivators, ultimately retarding p53-mediated growth arrest and apoptosis. In this study, we determined the molecular mechanism by which pVHL transactivates p53 on DNA damage and demonstrated that p53-related pVHL subtype mutants regulate tumorigenecity in VHL diseases.
- Published
- 2011
- Full Text
- View/download PDF
25. von Hippel–Lindau protein promotes Skp2 destabilization on DNA damage
- Author
-
In Young Hwang, Hong Duk Youn, Jae Seok Roe, Hwa Ryeon Kim, and Eun Jung Cho
- Subjects
Cancer Research ,Tumor suppressor gene ,DNA damage ,Apoptosis ,urologic and male genital diseases ,medicine.disease_cause ,S Phase ,Ubiquitin ,Genetics ,medicine ,SKP2 ,Humans ,Phosphorylation ,Carcinoma, Renal Cell ,S-Phase Kinase-Associated Proteins ,Molecular Biology ,biology ,DNA synthesis ,Protein Stability ,Ubiquitination ,HCT116 Cells ,Kidney Neoplasms ,female genital diseases and pregnancy complications ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,Kinetics ,HEK293 Cells ,Von Hippel-Lindau Tumor Suppressor Protein ,biology.protein ,Cancer research ,Carcinogenesis ,Proto-Oncogene Proteins c-akt ,Cyclin-Dependent Kinase Inhibitor p27 ,Cullin ,DNA Damage - Abstract
Germline mutations in the von Hippel-Lindau (VHL) tumor suppressor gene cause VHL disease, a rare and autosomal-dominant genetic syndrome. Because VHL protein (pVHL) is the master regulator of hypoxia-inducible factor alpha (HIFα), the most prominent feature of VHL disease is the deregulation of HIFα proteins. However, the precise mechanism by which the loss of pVHL function contributes to tumorigenesis is not fully understood. Here, we show that pVHL destabilizes the F-box protein Skp2, a chief component of Skp, Cullin, F-box-containing complex that promotes DNA synthesis in the S phase. The β-domain of pVHL interacts with Skp2, stimulating proteasome-dependent Skp2 degradation, but the destabilization of Skp2 does not depend on the E3 ubiquitin ligase activity of pVHL. Notably, the generation of DNA damage induces Skp2 degradation, which is attenuated by the suppression of endogenous pVHL expression. One possible mechanism of pVHL-dependent Skp2 degradation entails the antagonizing of Akt-mediated Skp2 phosphorylation, which maintains Skp2 stability. Reintroduction of VHL into VHL-null renal cell carcinoma (RCC) cells decreased Skp2 levels and restored DNA damage-dependent Skp2 degradation. These results identify the tumor suppressor function of pVHL in delaying the S-phase progression to inhibit cell proliferation on DNA damage. Clinically, this report explains as to why Skp2 accumulates abnormally in RCC tissues.
- Published
- 2011
- Full Text
- View/download PDF
26. Myogenic transcriptional activation of MyoD mediated by replication-independent histone deposition
- Author
-
Yong-Jin Yang, Yunkyoung Song, Jeung Whan Han, Jin Young Park, Hong Duk Youn, Eun Jung Cho, Jae Hyun Yang, and Ja Hwan Seol
- Subjects
Transcriptional Activation ,Chromatin Immunoprecipitation ,Immunoblotting ,Fluorescent Antibody Technique ,Cell Cycle Proteins ,Muscle Development ,Transfection ,MyoD ,Cell Line ,Histones ,Mice ,Histone H3 ,MyoD Protein ,RNA, Small Nuclear ,Animals ,Immunoprecipitation ,Histone code ,Histone Chaperones ,Epigenetics ,DNA Primers ,Multidisciplinary ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Biological Sciences ,Chromatin Assembly and Disassembly ,Microarray Analysis ,Molecular biology ,Chromatin ,Histone ,biology.protein ,RNA Interference ,Chromatin immunoprecipitation ,Transcription Factors - Abstract
In mammals, the canonical histone H3 and the variant H3.3 are assembled into chromatin through replication-coupled and replication-independent (RI) histone deposition pathways, respectively, to play distinct roles in chromatin function. H3.3 is largely associated with transcriptionally active regions via the activity of RI histone chaperone, HIRA. However, the precise role of the RI pathway and HIRA in active transcription and the mechanisms by which H3.3 affects gene activity are not known. In this study, we show that HIRA is an essential factor for muscle development by establishing MyoD activation in myotubes. HIRA and Asf1a, but not CHD1 or Asf1b, mediate H3.3 incorporation in the promoter and the critical upstream regulatory regions of the MyoD gene. HIRA and H3.3 are required for epigenetic transition into the more permissive chromatin structure for polymerase II recruitment to the promoter, regardless of transcription-associated covalent modification of histones. Our results suggest distinct epigenetic management of the master regulator with RI pathway components for cellular differentiation.
- Published
- 2010
- Full Text
- View/download PDF
27. Author response: Aurkb/PP1-mediated resetting of Oct4 during the cell cycle determines the identity of embryonic stem cells
- Author
-
Jong Hyuk Lee, Tae Wan Kim, Hye Ji Kim, Han-Teo Lee, Hyunsoo Kim, Hyonchol Jang, Min Young Suh, Sojung Kwak, Sang Eun Lee, Eun-Jung Cho, Jihoon Shin, Hong Duk Youn, and Sangho Lee
- Subjects
Identity (social science) ,Cell cycle ,Biology ,Embryonic stem cell ,Cell biology - Published
- 2016
- Full Text
- View/download PDF
28. Histone chaperones regulate histone exchange during transcription
- Author
-
Eun-Jung Cho, Hye Jin Kim, Jeung Whan Han, Ja-Hwan Seol, and Hong Duk Youn
- Subjects
Saccharomyces cerevisiae Proteins ,Transcription, Genetic ,Cell Cycle Proteins ,Saccharomyces cerevisiae ,Histone exchange ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Fungal Proteins ,Histones ,Histone H3 ,Histone H1 ,Histone H2A ,Histone methylation ,Histone code ,Gene Silencing ,Histone octamer ,Molecular Biology ,General Immunology and Microbiology ,General Neuroscience ,Nuclear Proteins ,Nucleosomes ,Repressor Proteins ,Biochemistry ,Histone methyltransferase ,Mutation ,RNA Polymerase II ,Gene Deletion ,Molecular Chaperones - Abstract
Transcription by RNA polymerase II is accompanied by dynamic changes in chromatin, including the eviction/deposition of nucleosomes or the covalent modification of histone subunits. This study examined the role of the histone H3/H4 chaperones, Asf1 and HIR, in histone mobility during transcription, with particular focus on the histone exchange pathway, using a dual histone expression system. The results showed that the exchange of H3/H4 normally occurs during transcription by the histone chaperones. Both Asf1 and HIR are important for histone deposition but have a different effect on histone exchange. While Asf1 mediated incorporation of external H3/H4 and renewal of pre-existing histones, HIR opposed it. The balance of two opposing activities might be an important mechanism for determining current chromatin states.
- Published
- 2007
- Full Text
- View/download PDF
29. Cabin1 Represses MEF2 Transcriptional Activity by Association with a Methyltransferase, SUV39H1
- Author
-
Hong Duk Youn, Eun Jung Cho, Hyungsoo Kim, Hyonchol Jang, and Doo Eun Choi
- Subjects
animal structures ,Transcription, Genetic ,Biology ,Biochemistry ,Cell Line ,Histone H3 ,Histone H1 ,Histone H2A ,Humans ,Histone code ,Protein Methyltransferases ,Amino Acids ,Promoter Regions, Genetic ,Molecular Biology ,Nuclear receptor co-repressor 2 ,MEF2 Transcription Factors ,EZH2 ,Histone-Lysine N-Methyltransferase ,Cell Biology ,Phosphoproteins ,musculoskeletal system ,Molecular biology ,Cell biology ,Myogenic Regulatory Factors ,Histone methyltransferase ,Histone Methyltransferases ,Calcium ,Heterochromatin protein 1 ,Protein Binding - Abstract
Myocyte enhancer factor 2 (MEF2) plays pivotal roles in various biological processes, and its transcriptional activity is regulated by histone acetylation/deacetylation enzymes in a calcium-dependent fashion. A calcineurin-binding protein 1 (Cabin1) has been shown to participate in repression of MEF2 by recruiting mSin3 and its associated histone deacetylases. Here, we report that histone methylation also takes a part in Cabin1-mediated repression of MEF2. Immunoprecipitate of Cabin1 complex can methylate histone H3 by association with SUV39H1. SUV39H1 increased Cabin1-mediated repression of MEF2 transcriptional activity in MEF2-targeting promoters. The SUV39H1 was revealed to bind to the 501-900-amino acid region of Cabin1, which was distinct from its histone deacetylase-recruiting domain. In addition, the Gal4-Cabin1-(501-900) alone repressed a constitutively active Gal4-tk-promoter, indicating that Cabin1 recruits SUV39H1 and represses transcriptional activity. Finally, both SUV39H1 and Cabin1 were shown to bind on the MEF2 target promoter in a calcium-dependent manner. Thus, Cabin1 recruits chromatin-modifying enzymes, both histone deacetylases and a histone methyltransferase, to repress MEF2 transcriptional activity.
- Published
- 2007
- Full Text
- View/download PDF
30. Dibutyryl cAMP stimulates the proliferation of SH-SY5Y human neuroblastoma cells by up-regulating Skp2 protein
- Author
-
Miran Seo, Yun Il Lee, Chin Ho Cho, Hong Duk Youn, Yong Sung Juhnn, and So Young Kim
- Subjects
Cancer Research ,medicine.medical_specialty ,Time Factors ,SH-SY5Y ,Biology ,S Phase ,Cell cycle phase ,Neuroblastoma ,Downregulation and upregulation ,Western blot ,Cyclins ,Internal medicine ,Tumor Cells, Cultured ,medicine ,Humans ,Kinase activity ,S-Phase Kinase-Associated Proteins ,Cell Proliferation ,Cyclin-Dependent Kinase Inhibitor Proteins ,Dose-Response Relationship, Drug ,medicine.diagnostic_test ,Cell growth ,Cyclin-Dependent Kinase 2 ,Cyclin-dependent kinase 2 ,G1 Phase ,General Medicine ,medicine.disease ,Up-Regulation ,Cell biology ,Endocrinology ,Bucladesine ,Oncology ,biology.protein ,Cyclin-Dependent Kinase Inhibitor p27 - Abstract
We previously found that the proliferation of SH-SY5Y neuroblastoma cells is stimulated when cAMP is up-regulated by stable expression of stimulatory G protein. Therefore, this study was performed to investigate the mechanism whereby cAMP stimulates the proliferation of SH-SY5Y cells. To investigate the effect of cAMP on cellular proliferation, SH-SY5Y neuroblastoma cells were treated with dibutyryl cAMP (dbcAMP), and then cell growth, thymidine incorporation and cell cycle phase distribution were analyzed. The expression and the activity of the molecules that regulate cell cycle progression were monitored by Western blot, RT-PCR, and kinase activity assay. Treatment with dbcAMP produced a biphasic effect on cellular proliferation; especially treatment with low concentration of dbcAMP (0.5 mM) showed a higher cellular proliferation rate and promoted G1/S transition in cell cycle. The dbcAMP (0.5 mM) treatment increased CDK2 activity, and it significantly decreased p27Kip1 expression with a decreased half-life of p27Kip1 protein. Moreover, dbcAMP (0.5 mM) increased the protein level and the stability of Skp2 with a concomitant decrease in its ubiquitination. cAMP up-regulates Skp2 protein by reducing its degradation probably through decreasing the ubiquitination of Skp2, which might result in accelerated degradation of p27Kip1, increase in CDK2 activity, and stimulation of SH-SY5Y cell proliferation in sequence.
- Published
- 2006
- Full Text
- View/download PDF
31. p53 Stabilization and Transactivation by a von Hippel-Lindau Protein
- Author
-
Sung Tae Kim, Jae Seok Roe, Hong Duk Youn, Soon Min Lee, Hyungsoo Kim, and Eun Jung Cho
- Subjects
Transcriptional Activation ,endocrine system diseases ,Elongin ,Active Transport, Cell Nucleus ,Apoptosis ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,law.invention ,Cancer syndrome ,Transactivation ,Proto-Oncogene Proteins c-mdm2 ,law ,Tumor Cells, Cultured ,medicine ,Humans ,Nuclear export signal ,neoplasms ,Transcription factor ,Molecular Biology ,Cell Nucleus ,Ubiquitin ,Lysine ,Tumor Suppressor Proteins ,Cell Cycle ,Acetylation ,Cell Biology ,Cell cycle ,medicine.disease ,female genital diseases and pregnancy complications ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Von Hippel-Lindau Tumor Suppressor Protein ,Cancer research ,Thermodynamics ,Suppressor ,Tumor Suppressor Protein p53 ,Carcinogenesis ,DNA Damage ,Protein Binding ,Transcription Factors - Abstract
von Hippel-Lindau (VHL) disease is a rare autosomal dominant cancer syndrome. Although hypoxia-inducible factor-alpha (HIFalpha) is a well-documented substrate of von Hippel-Lindau tumor suppressor protein (pVHL), it remains unclear whether the dysregulation of HIF is sufficient to account for de novo tumorigenesis in VHL-deleted cells. Here we found that pVHL directly associates with and stabilizes p53 by suppressing Mdm2-mediated ubiquitination and nuclear export of p53. Moreover, upon genotoxic stress, pVHL invoked an interaction between p53 and p300 and the acetylation of p53, which ultimately led to an increase in p53 transcriptional activity and p53-mediated cell cycle arrest and apoptosis. These results suggest that the tumor suppressor pVHL has an unexpected function to upregulate the tumor suppressor p53.
- Published
- 2006
- Full Text
- View/download PDF
32. CtBP represses p300-mediated transcriptional activation by direct association with its bromodomain
- Author
-
Jae Hwan Kim, Seong-Tae Kim, Hong Duk Youn, and Eun Jung Cho
- Subjects
Transcriptional Activation ,Molecular Sequence Data ,Cell Cycle Proteins ,P300-CBP Transcription Factors ,Biology ,Binding, Competitive ,Cell Line ,Histones ,Mice ,Histone H3 ,Acetyltransferases ,Structural Biology ,Histone H2A ,Animals ,Humans ,Histone code ,p300-CBP Transcription Factors ,Amino Acid Sequence ,Molecular Biology ,Histone Acetyltransferases ,Mice, Knockout ,Histone acetyltransferase ,NAD ,Phosphoproteins ,HDAC4 ,Protein Structure, Tertiary ,DNA-Binding Proteins ,Repressor Proteins ,Alcohol Oxidoreductases ,Biochemistry ,PCAF ,Histone methyltransferase ,Mutation ,biology.protein ,Sequence Alignment ,Protein Binding ,Transcription Factors - Abstract
Histone acetyltransferase coactivators bind to acetylated histones through their bromodomains and catalyze the acetylation of histone H3 and H4 tails for transcriptional activation. C-terminal binding protein (CtBP) serves as a transcriptional corepressor by recruiting histone deacetylases. However, the precise mechanism by which CtBP represses transcription has not been determined. In this study we found that CtBP1 directly associates with p300 by binding to the PXDLS motif in the bromodomain of p300. Moreover, CtBP1 blocks the accessibility of p300 to histones in an NADH-sensitive manner and thus represses p300-mediated histone acetylation and transcriptional activation. In addition, an NADH-nonresponsive, monomeric mutant, CtBP1 (G183V), was found to strongly repress p300-mediated transcriptional activation. Thus, the dissociation of NADH from CtBP1 leads to the repression of p300-driven general transcriptional activity by CtBP1. These results suggest a novel mechanism whereby CtBP1 serves as an energy-sensing repressor of histone acetyltransferase(s) and thus affects general transcription.
- Published
- 2005
- Full Text
- View/download PDF
33. mRNA Capping Enzyme Activity Is Coupled to an Early Transcription Elongation
- Author
-
Jeong Hwa Heo, Seok Ho Jeong, Hong Duk Youn, Seong-Tae Kim, Hye Jin Kim, Jeong Whan Han, Su Jin Jeong, Hyang Woo Lee, and Eun Jung Cho
- Subjects
RNA Caps ,Guanylyltransferase ,Saccharomyces cerevisiae Proteins ,Transcription, Genetic ,Antimetabolites ,Gene Expression ,RNA polymerase II ,Saccharomyces cerevisiae ,Biology ,Transcription (biology) ,Capping enzyme ,Gene Expression Regulation, Fungal ,Humans ,Promoter Regions, Genetic ,Uracil ,Molecular Biology ,Alleles ,Temperature ,Cell Biology ,DSIF ,Nucleotidyltransferases ,Protein Subunits ,Biochemistry ,Guanylyltransferase activity ,Mutation ,Transcription preinitiation complex ,Transcription factor II H ,biology.protein ,Cell Division - Abstract
The eukaryotic mRNAs produced by RNA polymerase II (Pol II) are capped with an inverted 7-methyl-guanosine (m7G) linked to the first residue of the mRNA (36). This event occurs by a series of three enzymatic reactions; The 5′ triphosphate end of the nascent RNA Pol II transcript is cleaved by 5′ RNA triphosphatase to produce the diphosphate end. RNA guanylyltransferase forms a covalent enzyme-GMP complex and subsequently caps the RNA substrate by adding a guanosine residue in a 5′-5′ triphosphate linkage. The cap is then methylated by RNA (guanine-7) methyltransferase (23, 39). In higher eukaryotes, a bifunctional monomeric polypeptide carries both the RNA triphosphatase and guanylyltransferase activities, while the capping enzyme from yeast is a complex of RNA triphosphatase and guanylyltransferase subunits (40). The polypeptides are encoded by the CET1 and CEG1 genes, respectively in Saccharomyces cerevisiae, and both are essential for the cell viability. Capping, the first mRNA modification, occurs by the time the transcript is only 25 to 30 nucleotides long in an early transcription phase. Such cotranscriptional capping is mediated by recruitment of capping enzyme machinery to the phosphorylated carboxy-terminal domain (CTD) of the largest subunit of Pol II (7, 15, 22, 51). The CTD of Pol II has an unusual structure with many heptapeptide repeats (YSPTSPS). The capping enzyme binds directly and specifically to the phosphorylated CTD of Pol II via the Ceg1 subunit (yeast) or the guanylyltransferase domain (metazoan). Furthermore, the guanylyltransferase activity of Ceg1 associated with phosphorylated CTD is allosterically regulated by both the Cet1 and phosphorylated CTD to ensure it has a coordinated capping activity (8). The mammalian capping enzyme is also allosterically regulated by an interaction with the phosphorylated CTD (16, 25). The CTD phosphorylated at serine 5 of the heptapeptide repeat appears to be important for the capping reaction because it stimulates the guanylyltransferase activity, although both serine 2 phosphorylation and serine 5 phosphorylation can mediate the protein interaction. An interesting extension to these findings was added by a chromatin immunoprecipitation assay, which provides in vivo evidence that the capping enzyme machinery interacts dynamically with Pol II during a transcription cycle (20, 35). The capping enzyme interacts with Pol II immediately after the serine 5 of CTD is phosphorylated. As serine 5 phosphorylation decreases in an early elongation phase, the capping enzyme dissociates from the transcription complex. Recently, data from several groups have raised the concept of “checkpoints” in transcription, especially in an early phase (summarized in reference 26). As operated during the cell cycle to ensure that each phase of the cycle is complete before the next one begins, checkpoint in early transcription is suggested to play a role in ensuring that only the properly modified RNA at the 5′ end is extended. The Pol II transcription is thus subjected to checkpoint control for the coordinated transcription with mRNA capping (6, 25, 26). Several transcription factors have been reported to play in this window. DSIF, a human homolog of the yeast transcription factor Spt4/Spt5, increases the pausing of Pol II and thus plays a role as a negative factor (42, 46, 49). Within this temporal and spatial interval, while Pol II with the hypophosphorylated CTD is paused at the promoter-proximal region, many factors are intended to target the capping enzymes to increase their recruitment or to enhance their catalytic activities. In addition to TFIIH, which creates a binding epitope for the capping enzyme on CTD by serine 5 phosphorylation, as described above, Spt5 itself interacts with the triphosphatase and guanylyltransferase components of the capping enzyme (21, 27, 44). In the case of human immunodeficiency virus (HIV), DSIF/Spt5-induced transcription arrest allows HIV-encoded Tat to interact with capping enzymes and to stimulate their catalytic activities (5, 6). Phosphorylation of Pol II CTD is critical for the transition to the elongation phase. At this step, the elongation factor P-TEFb, a DRB-sensitive protein kinase, phosphorylates the Pol II CTD and Spt5 (19, 29, 38). HIV Tat also interacts with P-TEFb in this step (29). In S. cerevisae, Ctk1 kinase complex and the Bur1 kinase complex facilitate the transcription elongation (18, 50). The phosphorylation of Pol II CTD is thus meant to lead to the formation of the processive transcription elongation complexes. According to recent reports, the capping enzyme in Schizosaccharomyces pombe interacts with Cdk9/Pch1, a yeast P-TEFb homolog (28). How does the capping enzyme fit into the complicated scheme to delineate it in the order of pausing, capping, and the reversion of pausing in the checkpoint model? If the elongation-competent transition does not occur until the RNA is capped and the capping is the major determinant to shift the Pol II status, it could be indicative that a capping enzyme plays a critical role in regulation of an early transcription in addition to its role in simple cap formation. To study whether the capping enzyme plays a key role in coordinating mRNA processing and transcription elongation, we used a well-characterized yeast system. Because Ceg1, a capping enzyme subunit, contributes to transcription via its typical cap formation activity, this study examined various ceg1 temperature-sensitive alleles to determine if there is any additional role in transcription elongation. Among them ceg1-63 displayed 6-azauracil (6AU) sensitivity and a defect in PUR5 induction by 6AU treatment. This study shows that transcription through the pause sites artificially inserted at the promoter-proximal region was severely inhibited in ceg1-63. We show that such an elongation defect was coupled to the reduced guanylyltransferase activity. However, it happened independently of the turnover of uncapped transcripts. These results indicate that the transcription ternary complexes are held at the promoter as long as their RNAs have not been properly capped. That is, capping enzyme plays a critical role in the promoter-proximal checkpoint window by reinforcing the checkpoint security circuit and probably by reversing the transcription arrest in time. This finding also strongly supports the transcription checkpoint model, in which an early transcription is tightly regulated.
- Published
- 2004
- Full Text
- View/download PDF
34. Nur77 Activated by Hypoxia-Inducible Factor-1α Overproduces Proopiomelanocortin in von Hippel-Lindau-Mutated Renal Cell Carcinoma
- Author
-
Ji-Woong Choi, Sang Chul Park, Jun O. Liu, Hong Duk Youn, and Gyeong Hoon Kang
- Subjects
Receptors, Steroid ,endocrine system ,Cancer Research ,Pro-Opiomelanocortin ,von Hippel-Lindau Disease ,Transcription, Genetic ,Nerve growth factor IB ,Ubiquitin-Protein Ligases ,Receptors, Cytoplasmic and Nuclear ,Adrenocorticotropic hormone ,Biology ,urologic and male genital diseases ,Proopiomelanocortin ,Transcription (biology) ,Nuclear Receptor Subfamily 4, Group A, Member 1 ,Tumor Cells, Cultured ,Humans ,Genes, Tumor Suppressor ,Binding site ,Promoter Regions, Genetic ,Receptor ,Carcinoma, Renal Cell ,Transcription factor ,DNA Primers ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Tumor Suppressor Proteins ,Cobalt ,Hypoxia-Inducible Factor 1, alpha Subunit ,Kidney Neoplasms ,DNA-Binding Proteins ,Amino Acid Substitution ,Oncology ,Hypoxia-inducible factors ,Von Hippel-Lindau Tumor Suppressor Protein ,Mutation ,biology.protein ,Cancer research ,Transcription Factors - Abstract
Mutation in the von Hippel-Lindau (VHL) protein associated with renal cell carcinoma causes hypoxia-inducible factor (HIF) to stabilize and consequently to induce various HIF-targeting proteins. In this study, we found that proopiomelanocortin (POMC), an adrenocorticotropic hormone precursor, is up-regulated constitutively in VHL-mutated renal cell carcinoma. A critical transcription factor responsible for POMC overproduction was identified as Nur77, a member of the orphan steroid receptor superfamily. Little is known about how VHL mutation leads to activation of Nur77. We report that Nur77 is directly regulated by HIF. We show that HIF-1α, but not HIF-2α, binds to a putative HIF responsive element in the Nur77 promoter, activating the expression of Nur77. Mutation or deletion of the HIF binding site in the Nur77 promoter abrogates activation of a luciferase reporter gene under the control of Nur77 promoter by HIF-1α. The treatment of Nur77 antisense oligonucleotide reduces POMC transcription under hypoxic conditions. We confirmed that Nur77 and POMC are up-regulated in VHL-mutated renal cell carcinoma. In this study, we provide the first molecular evidence that Nur77 activated by HIF under hypoxic conditions regulates production of the peptide hormone precursor POMC.
- Published
- 2004
- Full Text
- View/download PDF
35. Ctbp2-mediated β-catenin regulation is required for exit from pluripotency
- Author
-
Byung Hee Kang, Tae Wan Kim, Jihoon Shin, Sang Eun Lee, Jinmi Choi, Eun Jung Cho, Jong Hyuk Lee, Min Young Suh, Hong Duk Youn, Sojung Kwak, Jae Hwan Kim, and In Young Hwang
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Clinical Biochemistry ,Gene Expression ,Plasma protein binding ,Biology ,Biochemistry ,Models, Biological ,Cell Line ,03 medical and health sciences ,Mice ,Genes, Reporter ,Gene expression ,Animals ,Cell Self Renewal ,Nucleotide Motifs ,RNA, Small Interfering ,Molecular Biology ,Transcription factor ,Embryonic Stem Cells ,beta Catenin ,Genetics ,Gene knockdown ,Binding Sites ,Protein Stability ,Wnt signaling pathway ,Phosphoproteins ,Embryonic stem cell ,CTBP2 ,Cell biology ,DNA-Binding Proteins ,Alcohol Oxidoreductases ,030104 developmental biology ,Catenin ,Gene Knockdown Techniques ,embryonic structures ,Molecular Medicine ,Original Article ,Co-Repressor Proteins ,Protein Binding - Abstract
The canonical Wnt pathway is critical for embryonic stem cell (ESC) pluripotency and aberrant control of β-catenin leads to failure of exit from pluripotency and lineage commitments. Hence, maintaining the appropriate level of β-catenin is important for the decision to commit to the appropriate lineage. However, how β-catenin links to core transcription factors in ESCs remains elusive. C-terminal-binding protein (CtBP) in Drosophila is essential for Wnt-mediated target gene expression. In addition, Ctbp acts as an antagonist of β-catenin/TCF activation in mammals. Recently, Ctbp2, a core Oct4-binding protein in ESCs, has been reported to play a key role in ESC pluripotency. However, the significance of the connection between Ctbp2 and β-catenin with regard to ESC pluripotency remains elusive. Here, we demonstrate that C-terminal-binding protein 2 (Ctbp2) associates with major components of the β-catenin destruction complex and limits the accessibility of β-catenin to core transcription factors in undifferentiated ESCs. Ctbp2 knockdown leads to stabilization of β-catenin, which then interacts with core pluripotency-maintaining factors that are occupied by Ctbp2, leading to incomplete exit from pluripotency. These findings suggest a suppressive function for Ctbp2 in reducing the protein level of β-catenin, along with priming its position on core pluripotency genes to hinder β-catenin deposition, which is central to commitment to the appropriate lineage.
- Published
- 2017
36. Methylation and demethylation of DNA and histones in chromatin: the most complicated epigenetic marker
- Author
-
Hong Duk Youn
- Subjects
Genetic Markers ,0301 basic medicine ,Histone-modifying enzymes ,Clinical Biochemistry ,Biochemistry ,Epigenesis, Genetic ,Histones ,Cytosine ,03 medical and health sciences ,Epigenetics of physical exercise ,Neoplasms ,Histone methylation ,Animals ,Humans ,Molecular Biology ,RNA-Directed DNA Methylation ,Epigenomics ,Chemistry ,DNA Methylation ,Chromatin ,Cell biology ,DNA Demethylation ,Editorial ,030104 developmental biology ,DNA demethylation ,DNA methylation ,Molecular Medicine - Abstract
Methylation and demethylation of DNA and histones in chromatin: the most complicated epigenetic marker
- Published
- 2017
- Full Text
- View/download PDF
37. Ctbp2 Modulates NuRD-Mediated Deacetylation of H3K27 and Facilitates PRC2-Mediated H3K27me3 in Active Embryonic Stem Cell Genes During Exit from Pluripotency
- Author
-
Min Jueng Kang, Junil Kim, Jae Hwan Kim, Jihoon Shin, Sojung Kwak, Sang Eun Lee, Eun Jung Cho, Jeong Hwan Che, Byung Hee Kang, Hyonchol Jang, Dong Wook Han, Keun Soo Park, Jong Hyuk Lee, Hyunsoo Kim, Hong Duk Youn, Tae Wan Kim, Soon Min Lee, Eugene C. Yi, and Seon-Young Kim
- Subjects
macromolecular substances ,Cell Line ,Epigenesis, Genetic ,Histones ,Mice ,Nucleosome ,Gene silencing ,Animals ,Epigenetics ,Transcription factor ,reproductive and urinary physiology ,Genetics ,biology ,urogenital system ,Mouse Embryonic Stem Cells ,Cell Biology ,Chromatin Assembly and Disassembly ,Phosphoproteins ,Mi-2/NuRD complex ,Embryonic stem cell ,Cell biology ,Nucleosomes ,DNA-Binding Proteins ,Repressor Proteins ,Alcohol Oxidoreductases ,Histone ,embryonic structures ,biology.protein ,Molecular Medicine ,biological phenomena, cell phenomena, and immunity ,PRC2 ,Co-Repressor Proteins ,Developmental Biology ,Mi-2 Nucleosome Remodeling and Deacetylase Complex - Abstract
For cells to exit from pluripotency and commit to a lineage, the circuitry of a core transcription factor (CTF) network must be extinguished in an orderly manner through epigenetic modifications. However, how this choreographed epigenetic remodeling at active embryonic stem cell (ESC) genes occurs during differentiation is poorly understood. In this study, we demonstrate that C-terminal binding protein 2 (Ctbp2) regulates nucleosome remodeling and deacetylation (NuRD)-mediated deacetylation of H3K27 and facilitates recruitment of polycomb repressive complex 2 (PRC2)-mediated H3K27me3 in active ESC genes for exit from pluripotency during differentiation. By genomewide analysis, we found that Ctbp2 resides in active ESC genes and co-occupies regions with ESC CTFs in undifferentiated ESCs. Furthermore, ablation of Ctbp2 effects inappropriate gene silencing in ESCs by sustaining high levels of H3K27ac and impeding H3K27me3 in active ESC genes, thereby sustaining ESC maintenance during differentiation. Thus, Ctbp2 preoccupies regions in active genes with the NuRD complex in undifferentiated ESCs that are directed toward H3K27me3 by PRC2 to induce stable silencing, which is pivotal for natural lineage commitment. Stem Cells 2015;33:2442–2455
- Published
- 2014
38. Core Pluripotency Factors Directly Regulate Metabolism in Embryonic Stem Cell to Maintain Pluripotency
- Author
-
Hong Duk Youn, Hyonchol Jang, Eun Jung Cho, Hyun-Chul Kim, Doo Hyun Chung, Jihoon Shin, Byung Hee Kang, Tae Wan Kim, Sang Eun Lee, Yoon Kyung Jeon, and Jinmi Choi
- Subjects
Pluripotent Stem Cells ,Thyroid Hormones ,Rex1 ,Mice, SCID ,PKM2 ,Biology ,Mice ,Mice, Inbred NOD ,Hexokinase ,Animals ,Humans ,Glycolysis ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Membrane Proteins ,Cell Differentiation ,Cell Biology ,Metabolism ,Embryonic stem cell ,Cell biology ,Biochemistry ,Molecular Medicine ,Carrier Proteins ,Flux (metabolism) ,Leukemia inhibitory factor ,Octamer Transcription Factor-3 ,Developmental Biology - Abstract
Pluripotent stem cells (PSCs) have distinct metabolic properties that support their metabolic and energetic needs and affect their stemness. In particular, high glycolysis is critical for the generation and maintenance of PSCs. However, it is unknown how PSCs maintain and acquire this metabolic signature. In this study, we found that core pluripotency factors regulate glycolysis directly by controlling the expression of glycolytic enzymes. Specifically, Oct4 directly governs Hk2 and Pkm2, which are important glycolytic enzymes that determine the rate of glycolytic flux. The overexpression of Hk2 and Pkm2 sustains high levels of glycolysis during embryonic stem cell (ESC) differentiation. Moreover, the maintenance of high glycolysis levels by Hk2 and Pkm2 overexpression hampers differentiation and preserves the pluripotency of ESCs in the absence of leukemia inhibitory factor. Overall, our study identifies a direct molecular connection between core pluripotency factors and ESC metabolic signatures and demonstrates the significance of metabolism in cell fate determination. Stem Cells 2015;33:2699–2711
- Published
- 2014
39. ATP-citrate lyase regulates cellular senescence via an AMPK- and p53-dependent pathway
- Author
-
Ji-Eun Lee, Hyonchol Jang, Jong Hyuk Lee, Jinmi Choi, Tae Wan Kim, Soon Min Lee, Hong Duk Youn, and Eun Jung Cho
- Subjects
ATP citrate lyase ,Carcinogenesis ,AMP-Activated Protein Kinases ,Biochemistry ,Gene Expression Regulation, Enzymologic ,chemistry.chemical_compound ,Cytosol ,Acetyl Coenzyme A ,Neoplasms ,Citrate synthase ,Animals ,Humans ,Protein kinase A ,Molecular Biology ,Fatty acid synthesis ,Cellular Senescence ,Gene knockdown ,biology ,Chemistry ,Acetyl-CoA ,AMPK ,Cell Biology ,Cell biology ,Rats ,HEK293 Cells ,Gene Knockdown Techniques ,Cancer cell ,biology.protein ,ATP Citrate (pro-S)-Lyase ,Tumor Suppressor Protein p53 ,Signal Transduction - Abstract
ATP citrate lyase (ACLY) is a key enzyme that is involved in de novo lipogenesis by catalyzing conversion of cytosolic citrate into acetyl CoA and oxaloacetate. Up-regulation of ACLY in various types of tumors enhances fatty acid synthesis and supplies excess acetyl CoA for histone acetylation. However, there is evidence that its enzymatic activity alone is insufficient to explain ACLY silencing-mediated growth arrest in tumor cells. In this study, we found that ACLY knockdown in primary human cells triggers cellular senescence and activation of tumor suppressor p53. Provision of acetyl CoA to ACLY knockdown cells did not alleviate ACLY silencing-induced p53 activation, suggesting an independent role for ACLY activity. Instead, ACLY physically interacted with the catalytic subunit of AMP-activated protein kinase (AMPK) and inhibited AMPK activity. The activation of AMPK under ACLY knockdown conditions may lead to p53 activation, ultimately leading to cellular senescence. In cancer cells, ACLY silencing-induced p53 activation facilitated DNA damage-induced cell death. Taken together, our results suggest a novel function of ACLY in cellular senescence and tumorigenesis.
- Published
- 2014
40. Cabin1 Represses MEF2-Dependent Nur77 Expression and T Cell Apoptosis by Controlling Association of Histone Deacetylases and Acetylases with MEF2
- Author
-
Jun O. Liu and Hong Duk Youn
- Subjects
Receptors, Steroid ,T-Lymphocytes ,Histone Deacetylase 2 ,Receptors, Cytoplasmic and Nuclear ,Apoptosis ,Histone Deacetylase 1 ,Jurkat Cells ,Histone methylation ,Nuclear Receptor Subfamily 4, Group A, Member 1 ,Tumor Cells, Cultured ,Immunology and Allergy ,Promoter Regions, Genetic ,Nuclear receptor co-repressor 1 ,Nuclear receptor co-repressor 2 ,Histone Acetyltransferases ,Histone deacetylase 5 ,MEF2 Transcription Factors ,Calcineurin ,Nuclear Proteins ,musculoskeletal system ,DNA-Binding Proteins ,Sin3 Histone Deacetylase and Corepressor Complex ,Infectious Diseases ,Myogenic Regulatory Factors ,Histone methyltransferase ,embryonic structures ,cardiovascular system ,tissues ,Transcriptional Activation ,Saccharomyces cerevisiae Proteins ,animal structures ,Immunology ,Biology ,Binding, Competitive ,Histone Deacetylases ,Sp3 transcription factor ,Acetyltransferases ,Histone H2A ,Humans ,Calcium Signaling ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Binding Sites ,DNA ,Phosphoproteins ,Protein Structure, Tertiary ,Repressor Proteins ,Cancer research ,Trans-Activators ,Transcription Factors - Abstract
TCR signaling leading to thymocyte apoptosis is mediated through the expression of the Nur77 family of orphan nuclear receptors. MEF2 has been shown to be the major transcription factor responsible for calcium-dependent Nur77 transcription. Cabin1 was recently identified as a transcriptional repressor of MEF2, which can be released from MEF2 in a calcium-dependent fashion. The molecular basis of repression of MEF2 by Cabin1, however, has remained unknown. We report that Cabin1 represses MEF2 by two distinct mechanisms. Cabin1 recruits mSin3 and its associated histone deacetylases 1 and 2; Cabin1 also competes with p300 for binding to MEF2. Thus, activation of MEF2 and the consequent transcription of Nur77 are controlled by the association of MEF2 with the histone deacetylases via the calcium-dependent repressor Cabin1.
- Published
- 2000
- Full Text
- View/download PDF
41. Apoptosis of T Cells Mediated by Ca2+-Induced Release of the Transcription Factor MEF2
- Author
-
Ron Prywes, Hong Duk Youn, Jun O. Liu, and Luo Sun
- Subjects
Receptors, Steroid ,animal structures ,Transcription, Genetic ,Nerve growth factor IB ,Calmodulin ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Gene Expression ,Receptors, Cytoplasmic and Nuclear ,Apoptosis ,Cell Line ,Jurkat Cells ,Genes, Reporter ,Two-Hybrid System Techniques ,Nuclear Receptor Subfamily 4, Group A, Member 1 ,Humans ,Calcium Signaling ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Calcium signaling ,Multidisciplinary ,biology ,MEF2 Transcription Factors ,Calcineurin ,T-cell receptor ,Phosphoproteins ,musculoskeletal system ,Cell biology ,DNA-Binding Proteins ,Thymocyte ,Myogenic Regulatory Factors ,embryonic structures ,cardiovascular system ,Cancer research ,biology.protein ,Calcium ,Signal transduction ,tissues ,Transcription Factors - Abstract
T cell receptor (TCR)–induced apoptosis of thymocytes is mediated by calcium-dependent expression of the steroid receptors Nur77 and Nor1. Nur77 expression is controlled by the transcription factor myocyte enhancer factor 2 (MEF2), but how MEF2 is activated by calcium signaling is still obscure. Cabin1, a calcineurin inhibitor, was found to regulate MEF2. MEF2 was normally sequestered by Cabin1 in a transcriptionally inactive state. TCR engagement led to an increase in intracellular calcium concentration and the dissociation of MEF2 from Cabin1, as a result of competitive binding of activated calmodulin to Cabin1. The interplay between Cabin1, MEF2, and calmodulin defines a distinct signaling pathway from the TCR to the Nur77 promoter during T cell apoptosis.
- Published
- 1999
- Full Text
- View/download PDF
42. Oct4 resetting by Aurkb–PP1 cell cycle axis determines the identity of mouse embryonic stem cells
- Author
-
Jihoon Shin and Hong Duk Youn
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Embryonic stem cells ,M Phase Cell Cycle Checkpoints ,Somatic cell ,Biology ,Cell cycle ,Biochemistry ,03 medical and health sciences ,Mice ,Protein Phosphatase 1 ,Animals ,Aurora Kinase B ,Phosphorylation ,Induced pluripotent stem cell ,Molecular Biology ,Transcription factor ,reproductive and urinary physiology ,030102 biochemistry & molecular biology ,Oct4 resetting ,fungi ,Mouse Embryonic Stem Cells ,General Medicine ,PP1 ,Cellular Reprogramming ,Embryonic stem cell ,G1 Phase Cell Cycle Checkpoints ,Chromatin ,Cell biology ,Aurkb ,embryonic structures ,Perspective ,Mutation ,biological phenomena, cell phenomena, and immunity ,Reprogramming ,Octamer Transcription Factor-3 - Abstract
In embryonic stem cells (ESCs), cell cycle regulation is deeply connected to pluripotency. Especially, core transcription factors (CTFs) which are essential to maintaining the pluripotency transcription programs should be reset during M/G1 transition. However, it remains unknown about how CTFs are governed during cell cycle progression. Here, we describe that the regulation of Oct4 by Aurora kinase b (Aurkb)/protein phosphatase 1 (PP1) axis during the cell cycle is important for resetting Oct4 to pluripotency and cell cycle related target genes in determining the identity of ESCs. Aurkb starts to phosphorylate Oct4(S229) at the onset of G2/M phase, inducing the dissociation of Oct4 from chromatin, whereas PP1 binds Oct4 and dephosphorylates Oct4(S229) during M/G1 transition, which resets Oct4-driven transcription for pluripotency and the cell cycle. Furthermore, Aurkb phosphormimetic and PP1 binding-deficient mutations in Oct4 disrupt the pluripotent cell cycle, lead to the loss of pluripotency in ESCs, and decrease the efficiency of somatic cell reprogramming. Based on our findings, we suggest that the cell cycle is directly linked to pluripotency programs in ESCs. [BMB Reports 2016; 49(10): 527-528].
- Published
- 2016
43. Streptomyces seoulensis sp. nov
- Author
-
Hong Duk Youn, Min-Young Kim, Yung Chil Hah, Hong Kum Lee, Sa-Ouk Kang, Yang I.N. Yim, and Jongsik Chun
- Subjects
DNA, Bacterial ,Molecular Sequence Data ,Immunology ,Streptomyces seoulensis ,DNA, Ribosomal ,Microbiology ,Streptomyces ,Species Specificity ,Phylogenetics ,RNA, Ribosomal, 16S ,Botany ,Phylogeny ,Soil Microbiology ,Korea ,biology ,Computers ,Streptomycetaceae ,Ribosomal RNA ,16S ribosomal RNA ,biology.organism_classification ,RNA, Bacterial ,Phenotype ,Genes, Bacterial ,Taxonomy (biology) ,Actinomycetales - Abstract
The taxonomic position of an actinomycete strain isolated from Korean soil was examined by a polyphasic approach. The isolate, designated IMSNU-1, was clearly assigned to the genus Streptomyces on the basis of morphological and chemotaxonomic data. The test strain was the subject of a probabilistic identification study using the identification matrices generated by Langham et al. (J. Gen. Microbiol. 135:121-133, 1989) and found to be marginally close to clusters 19 and 39. An almost complete 16S rRNA gene (rDNA) sequence was obtained for the test strain and compared with those of representative streptomycetes. 16S rDNA sequence data not only support the strain's membership in the genus Streptomyces but also provide strong evidence that our isolate is genealogically distant from representatives of clusters 19 and 39, forming a separate phyletic line in a clade encompassed by streptomycetes. It is therefore proposed from the polyphasic evidence that strain IMSNU-1 be classified in the genus Streptomyces as Streptomyces seoulensis sp. nov.
- Published
- 1997
- Full Text
- View/download PDF
44. Menin mediates epigenetic regulation via histone H3 lysine 9 methylation
- Author
-
E. J. Cho, Ho-Young Lee, C. H. Jo, T. Y. Song, J. H. Yang, Y. Song, Jinseon Lee, Jong Woo Park, Y. N. Kim, Augustine M.K. Choi, Yong-Jin Yang, Hyonchol Jang, Hong Duk Youn, Jeung Whan Han, J. Lim, and Seong-Tae Kim
- Subjects
Cancer Research ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system ,endocrine system diseases ,tumor suppressor ,Immunology ,Biology ,Methylation ,SUV39H1 ,Chromatin remodeling ,Epigenesis, Genetic ,menin ,Histones ,Cellular and Molecular Neuroscience ,Histone H3 ,Mice ,Proto-Oncogene Proteins ,Histone methylation ,Animals ,Humans ,MEN1 ,Epigenetics ,histone methylation ,Homeodomain Proteins ,Lysine ,Cell Biology ,Methyltransferases ,Histone H3-K4 methylation ,Fibroblasts ,Chromatin Assembly and Disassembly ,Repressor Proteins ,Protein Transport ,Cell Transformation, Neoplastic ,HEK293 Cells ,Histone methyltransferase ,Mutation ,Cancer research ,Original Article ,Chromatin immunoprecipitation ,Signal Transduction - Abstract
Menin, encoded by the multiple endocrine neoplasia type 1 (MEN1) gene, is a tumor suppressor that leads to multiple endocrine tumors upon loss of its function. Menin functions as a transcriptional activator by tethering MLL complex to mediate histone H3 K4 methylation. It also functions as a repressor. However, the molecular mechanism of how menin contributes to the opposite outcome in gene expression is largely unknown. Here, we investigated the role of menin in the epigenetic regulation of transcription mediated by histone covalent modification. We show that the global methylation level of histone H3 K9, as well as H3 K4, was decreased in Men1(-/-) MEF cells. Consistently, menin was able to interact with the suppressor of variegation 3-9 homolog family protein, SUV39H1, to mediate H3 K9 methylation. This interaction decreased when patient-derived MEN1 mutation was introduced into the SUV39H1-interaction domain. We show that menin mediated different chromatin changes depending on target genes. Chromatin immunoprecipitation studies showed that menin directly associated with the GBX2 promoter and menin-dependent recruitment of SUV39H1 was essential for chromatin remodeling and transcriptional regulation. These results provide a molecular basis of how menin functions as a transcriptional repressor and suggest that menin-dependent integration of H3 K9 methylation might play an important role in preventing tumors. Cell Death and Disease (2013) 4, e583; doi:10.1038/cddis.2013.98; published online 11 April 2013
- Published
- 2013
45. Role of laccase in lignin degradation by white-rot fungi
- Author
-
Hong-Duk Youn, Yung Chil Hah, and Sa-Ouk Kang
- Subjects
Genetics ,Molecular Biology ,Microbiology - Published
- 1995
- Full Text
- View/download PDF
46. A phosphorylation pattern-recognizing antibody specifically reacts to RNA polymerase II bound to exons
- Author
-
Won Jun Yang, Hyori Kim, Do Been Hwang, Junho Chung, Yujean Lee, Sunyoung Park, S.-C. Yoon, Jong Hyuk Lee, Hwa K. Lee, Hong Duk Youn, Aerin Yoon, Min S. Kim, and J. K. Han
- Subjects
0301 basic medicine ,Chromatin Immunoprecipitation ,Clinical Biochemistry ,RNA polymerase II ,Biochemistry ,Antibodies ,03 medical and health sciences ,chemistry.chemical_compound ,Exon ,Protein biosynthesis ,Humans ,Phosphorylation ,Molecular Biology ,Gene ,biology ,Intron ,Exons ,Molecular biology ,Synthetic antibody ,030104 developmental biology ,HEK293 Cells ,chemistry ,Phosphoserine ,biology.protein ,Molecular Medicine ,Original Article ,RNA Polymerase II ,DNA ,HeLa Cells ,Protein Binding - Abstract
The C-terminal domain of RNA polymerase II is an unusual series of repeated residues appended to the C-terminus of the largest subunit and serves as a flexible binding scaffold for numerous nuclear factors. The binding of these factors is determined by the phosphorylation patterns on the repeats in the domain. In this study, we generated a synthetic antibody library by replacing the third heavy chain complementarity-determining region of an anti-HER2 (human epidermal growth factor receptor 2) antibody (trastuzumab) with artificial sequences of 7–18 amino-acid residues. From this library, antibodies were selected that were specific to serine phosphopeptides that represent typical phosphorylation patterns on the functional unit (YSPTSPS)2 of the RNA polymerase II C-terminal domain (CTD). Antibody clones pCTD-1stS2 and pCTD-2ndS2 showed specificity for peptides with phosphoserine at the second residues of the first or second heptamer repeat, respectively. Additional clones specifically reacted to peptides with phosphoserine at the fifth serine of the first repeat (pCTD-1stS5), the seventh residue of the first repeat and fifth residue of the second repeat (pCTD-S7S5) or the seventh residue of either the first or second repeat (pCTD-S7). All of these antibody clones successfully reacted to RNA polymerase II in immunoblot analysis. Interestingly, pCTD-2ndS2 precipitated predominately RNA polymerase II from the exonic regions of genes in genome-wide chromatin immunoprecipitation sequencing analysis, which suggests that the phosphoserine at the second residue of the second repeat of the functional unit (YSPTSPS)2 is a mediator of exon definition. Antibodies that bind to an enzyme that allows genes to make the proteins they encode could prove a valuable research tool. The ‘RNA polymerase II’ enzyme copies a gene into messenger RNAs – molecules that carry genetic information from the cell nucleus to direct protein synthesis out in the cytoplasm. The enzyme has an unusual pattern of amino acids carrying phosphate groups at one end of its protein chain. Hyori Kim at the University of Ulsan in South Korea, with Junho Chung and co-workers at Seoul National University, created a series of antibodies that selectively bind to these ‘phosphorylated’ regions of the enzyme depending on the phosphorylation pattern. The antibodies are being used to explore how different phosphorylation patterns control the binding of this vital enzyme to molecules required for gene activity, and to specific sites on DNA like exon and intron.
- Published
- 2016
47. Differential regulation of the histone chaperone HIRA during muscle cell differentiation by a phosphorylation switch
- Author
-
Eun Jung Cho, Jeung Whan Han, Tae Yang Song, Ilang Song, Jinyoung Park, Hong Duk Youn, Kwan Young Jung, Suji Hong, Jae-Hoon Kim, Han Young Lee, Jae Hyun Yang, and Chanhee Jo
- Subjects
Transcriptional Activation ,0301 basic medicine ,Cellular differentiation ,Clinical Biochemistry ,Cell Cycle Proteins ,Muscle Development ,MyoD ,Biochemistry ,Cell Line ,Histones ,Myoblasts ,Mice ,03 medical and health sciences ,medicine ,Animals ,Humans ,Histone Chaperones ,Phosphorylation ,Molecular Biology ,Regulation of gene expression ,biology ,Myogenesis ,Muscle cell differentiation ,Gene Expression Regulation, Developmental ,Skeletal muscle ,Cell Differentiation ,Cell biology ,030104 developmental biology ,Histone ,medicine.anatomical_structure ,biology.protein ,Molecular Medicine ,Original Article ,Proto-Oncogene Proteins c-akt ,Transcription Factors - Abstract
Replication-independent incorporation of variant histone H3.3 has a profound impact on chromatin function and numerous cellular processes, including the differentiation of muscle cells. The histone chaperone HIRA and H3.3 have essential roles in MyoD regulation during myoblast differentiation. However, the precise mechanism that determines the onset of H3.3 deposition in response to differentiation signals is unclear. Here we show that HIRA is phosphorylated by Akt kinase, an important signaling modulator in muscle cells. By generating a phosphospecific antibody, we found that a significant amount of HIRA was phosphorylated in myoblasts. The phosphorylation level of HIRA and the occupancy of phosphorylated protein on muscle genes gradually decreased during cellular differentiation. Remarkably, the forced expression of the phosphomimic form of HIRA resulted in reduced H3.3 deposition and suppressed the activation of muscle genes in myotubes. Our data show that HIRA phosphorylation limits the expression of myogenic genes, while the dephosphorylation of HIRA is required for proficient H3.3 deposition and gene activation, demonstrating that the phosphorylation switch is exploited to modulate HIRA/H3.3-mediated muscle gene regulation during myogenesis.
- Published
- 2016
- Full Text
- View/download PDF
48. Single electron transfer by an extracellular laccase from the white-rot fungus Pleurotus ostreatus
- Author
-
Young-Hoon Han, Sa-Ouk Kang, Jin-Soo Maeng, Yung Chil Hah, Gajin Jeong, Hong Duk Youn, In-Beom Jeong, and Kyu-Jung Kim
- Subjects
Antigens, Fungal ,Hot Temperature ,Immunoblotting ,Molecular Sequence Data ,Size-exclusion chromatography ,Microbiology ,Substrate Specificity ,Electron Transport ,Polyporaceae ,Ferulic acid ,chemistry.chemical_compound ,Metalloproteins ,Organic chemistry ,Amino Acid Sequence ,Polyacrylamide gel electrophoresis ,Laccase ,Sequence Homology, Amino Acid ,biology ,Molecular mass ,Electron Spin Resonance Spectroscopy ,Active site ,Hydrogen-Ion Concentration ,Syringic acid ,biology.organism_classification ,chemistry ,biology.protein ,Pleurotus ostreatus ,Oxidoreductases ,Sequence Analysis ,Copper ,Nuclear chemistry - Abstract
Two different bands with laccase activity were obtained after nondenaturing PAGE of the culture filtrate of Pleurotus ostreatus. Immunoblot analysis revealed that antisera raised against laccase I were not reactive to laccase II. Laccase I, which exhibited faster mobility on nondenaturing polyacrylamide gel, was purified 42.9-fold with an overall yield of 10.8%. Gel filtration and SDS-PAGE revealed that laccase I is a single polypeptide with a molecular mass of approximately 64 kDa. Laccase I contained 12.5% carbohydrate by weight and 3.9 mol copper (mol protein)-1. The absorption spectrum of laccase I showed a type 1 signal at 605 nm and EPR spectra showed that the parameters of the type 1 and type 2 Cu signals were g parallel = 2.197 and A parallel = 0.009 cm-1, and g parallel = 2.263 and A parallel = 0.0176 cm-1, respectively. The data obtained from the pH profiles suggested that two ionization groups, whose pKa values were 5.60-5.70 and 6.70-6.85, may play an important role in the active site of laccase I as the ligand of copper metal. The optimal pH and temperature for the activity of laccase I were 6.0-6.5 and 30-35 degrees C, respectively. The enzyme had affinity for various lignin-related phenolic compounds: the Km values for ferulic acid and syringic acid were 48 and 89 microM, respectively. EPR spectroscopic study of the action of laccase I on 3,5-dimethoxy-5-hydroxyacetophenone indicated that this enzyme catalyses single electron transfer with the formation of the phenoxy radical as an intermediate.
- Published
- 1995
- Full Text
- View/download PDF
49. CHK2 kinase promotes pre-mRNA splicing via phosphorylating CDK11p110
- Author
-
Hee-Hyun Choi, Jill M. Lahti, Keejung Yoon, Sung Yun Jung, Hyun-Kyung Choi, Eun-Jung Cho, Seong-Tae Kim, Judith Hyle, Jun Qin, Beom-Jun Kim, and Hong Duk Youn
- Subjects
Cancer Research ,animal structures ,RNA Splicing ,P70-S6 Kinase 1 ,Mitogen-activated protein kinase kinase ,environment and public health ,Article ,MAP2K7 ,Protein Interaction Mapping ,Genetics ,RNA Precursors ,Humans ,ASK1 ,Amino Acid Sequence ,RNA, Messenger ,Kinase activity ,Phosphorylation ,Molecular Biology ,biology ,MAP kinase kinase kinase ,Cyclin-dependent kinase 2 ,Molecular biology ,Cyclin-Dependent Kinases ,enzymes and coenzymes (carbohydrates) ,Checkpoint Kinase 2 ,HEK293 Cells ,biology.protein ,Cyclin-dependent kinase 9 ,biological phenomena, cell phenomena, and immunity ,Protein Multimerization ,HT29 Cells ,Protein Processing, Post-Translational ,DNA Damage - Abstract
Checkpoint kinase 2 (CHK2) kinase is a key mediator in many cellular responses to genotoxic stresses, including ionizing radiation (IR) and topoisomerase inhibitors. Upon IR, CHK2 is activated by ataxia telangiectasia mutated kinase and regulates the S-phase and G1-S checkpoints, apoptosis and DNA repair by phosphorylating downstream target proteins, such as p53 and Brca1. In addition, CHK2 is thought to be a multi-organ cancer susceptibility gene. In this study, we used a tandem affinity purification strategy to identify proteins that interact with CHK2 kinase. Cyclin-dependent kinase 11 (CDK11)(p110) kinase, implicated in pre-mRNA splicing and transcription, was identified as a CHK2-interacting protein. CHK2 kinase phosphorylated CDK11(p110) on serine 737 in vitro. Unexpectedly, CHK2 kinase constitutively phosphorylated CDK11(p110) in a DNA damage-independent manner. At a molecular level, CDK11(p110) phosphorylation was required for homodimerization without affecting its kinase activity. Overexpression of CHK2 promoted pre-mRNA splicing. Conversely, CHK2 depletion decreased endogenous splicing activity. Mutation of the phosphorylation site in CDK11(p110) to alanine abrogated its splicing-activating activity. These results provide the first evidence that CHK2 kinase promotes pre-mRNA splicing via phosphorylating CDK11(p110).
- Published
- 2012
50. The tumor suppressor, parafibromin, mediates histone H3 K9 methylation for cyclin D1 repression
- Author
-
Yong-Jin Yang, Jeung Whan Han, Hong Duk Youn, and Eun Jung Cho
- Subjects
Binding Sites ,biology ,Transcription, Genetic ,Tumor Suppressor Proteins ,Parafibromin ,Repressor ,Methylation ,Histone-Lysine N-Methyltransferase ,Methyltransferases ,Gene Regulation, Chromatin and Epigenetics ,Cell Line ,Histones ,Repressor Proteins ,Histone H3 ,Histone ,Cyclin D1 ,Transcription (biology) ,Histone methyltransferase ,Genetics ,biology.protein ,Cancer research ,Histone Methyltransferases ,Humans - Abstract
Parafibromin, a component of the RNA polymerase II-associated PAF1 complex, is a tumor suppressor linked to hyperparathyroidism-jaw tumor syndrome and sporadic parathyroid carcinoma. Parafibromin induces cell cycle arrest by repressing cyclin D1 via an unknown mechanism. Here, we show that parafibromin interacts with the histone methyltransferase, SUV39H1, and functions as a transcriptional repressor. The central region (128-227 amino acids) of parafibromin is important for both the interaction with SUV39H1 and transcriptional repression. Parafibromin associated with the promoter and coding regions of cyclin D1 and was required for the recruitment of SUV39H1 and the induction of H3 K9 methylation but not H3 K4 methylation. RNA interference analysis showed that SUV39H1 was critical for cyclin D1 repression. These data suggest that parafibromin plays an unexpected role as a repressor in addition to its widely known activity associated with transcriptional activation. Parafibromin as a part of the PAF1 complex might downregulate cyclin D1 expression by integrating repressive H3 K9 methylation during transcription.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.