36 results on '"Lin-Feng, Chen"'
Search Results
2. Brd4 regulates NLRC4 inflammasome activation by facilitating IRF8-mediated transcription of Naips
- Author
-
Yan Bao, James M. Slauch, Xiangming Hu, Lin Feng Chen, Xiao Dong Yang, Guo Li, and Xingchen Dong
- Subjects
Salmonella typhimurium ,Chromatin or Epigenetics ,Transcription, Genetic ,Inflammasomes ,Immunology ,Caspase 1 ,Inflammation ,Biology ,Microbiology ,Models, Biological ,Article ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Cell Signaling ,NLRC4 ,Proto-Oncogene Proteins ,medicine ,Pyroptosis ,Animals ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Innate immune system ,Base Sequence ,Macrophages ,Calcium-Binding Proteins ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,Inflammasome ,Cell Biology ,Phosphate-Binding Proteins ,Neuronal Apoptosis-Inhibitory Protein ,Cell biology ,CARD Signaling Adaptor Proteins ,030220 oncology & carcinogenesis ,Interferon Regulatory Factors ,Trans-Activators ,CpG Islands ,IRF8 ,medicine.symptom ,Apoptosis Regulatory Proteins ,medicine.drug ,Protein Binding ,Transcription Factors - Abstract
Dong et al. show that the epigenetic regulator Brd4 forms a complex with transcription factors IRF8/PU.1 to activate the transcription of NLRC4 inflammasome component Naips in macrophages. Brd4-dependent inflammasome activation protects mice from Salmonella infection via cytokine release and pyroptosis., NLRC4 inflammasome activation and the subsequent maturation of IL-1β and IL-18 are critical for protection against infection by bacterial pathogens. The epigenetic regulator Brd4 has emerged as a key player in inflammation by regulating the expression of inflammatory cytokines. However, whether Brd4 has any role in inflammasome activation remains undetermined. Here, we demonstrated that Brd4 is an important regulator of NLRC4 inflammasome activation in response to Salmonella typhimurium infection. Brd4-deficient bone marrow–derived macrophages (BMDMs) displayed impaired caspase-1 activation, ASC oligomerization, IL-1β maturation, gasdermin-D cleavage, and pyroptosis in response to S. typhimurium infection. RNA sequencing and RT-PCR results revealed that the transcription of Naips was decreased in Brd4-deficient BMDMs. Brd4 formed a complex with IRF8/PU.1 and bound to the IRF8 and PU.1 binding motifs on the promoters of Naips to maintain the expression of Naips. Furthermore, myeloid lineage–specific Brd4 conditional knockout mice were more susceptible to S. typhimurium infection with increased mortality, bacterial loads, and tissue damage; impaired inflammasome-dependent cytokine production; and pyroptosis. Our studies identify a novel function of Brd4 in innate immunity by controlling inflammasome-mediated cytokine release and pyroptosis to effectively battle S. typhimurium infection.
- Published
- 2021
3. Measuring NF-κB Phosphorylation and Acetylation
- Author
-
Nikita Tushar Modi and Lin Feng Chen
- Subjects
chemistry.chemical_compound ,chemistry ,Acetylation ,In vivo ,Protein subunit ,Phosphorylation ,NF-κB ,Methylation ,Transcription factor ,In vitro ,Cell biology - Abstract
Posttranslational modifications of NF-κB, including phosphorylation, acetylation, and methylation, have emerged as important regulatory mechanisms to control the transcriptional outcomes of this important transcription factor. These modifications work independently, sequentially or in combination to modulate the diverse biological functions of NF-κB in cancer and inflammatory response. Here, we describe some experimental methods to detect the in vitro and in vivo phosphorylation and acetylation of NF-κB, specifically focusing on the RelA subunit of NF-κB. These methods include labeling the phospho- or acetyl- groups with radioisotopes in vitro and immunoblotting with site-specific anti-phospho-serine or acetyl-lysine antibodies in culture cells and tissue samples.
- Published
- 2021
4. H. pylori infection confers resistance to apoptosis via Brd4-dependent BIRC3 eRNA synthesis
- Author
-
Xingchen Dong, Xiangming Hu, Yana Zavros, Meihua Chen, Donald Sheppard, Ruichuan Chen, Lin Feng Chen, Yanheng Chen, Richard M. Peek, and Jayati Chakrabarti
- Subjects
Cell death ,Cancer Research ,Programmed cell death ,BRD4 ,Immunology ,Mutant ,Apoptosis ,Cell Cycle Proteins ,Inflammation ,Article ,Helicobacter Infections ,Cellular and Molecular Neuroscience ,Bacterial Proteins ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Humans ,lcsh:QH573-671 ,Enhancer ,Antigens, Bacterial ,Helicobacter pylori ,Caspase 3 ,lcsh:Cytology ,Chemistry ,Stomach ,Nuclear Proteins ,Cancer ,Epithelial Cells ,Cell Biology ,bacterial infections and mycoses ,medicine.disease ,Baculoviral IAP Repeat-Containing 3 Protein ,Enhancer Elements, Genetic ,Gastric Mucosa ,Cancer research ,RNA, Long Noncoding ,medicine.symptom ,Function (biology) ,Transcription Factors - Abstract
H. pylori infection is one of the leading causes of gastric cancer and the pathogenicity of H. pylori infection is associated with its ability to induce chronic inflammation and apoptosis resistance. While H. pylori infection-induced expression of pro-inflammatory cytokines for chronic inflammation is well studied, the molecular mechanism underlying the apoptosis resistance in infected cells is not well understood. In this study, we demonstrated that H. pylori infection-induced apoptosis resistance in gastric epithelial cells triggered by Raptinal, a drug that directly activates caspase-3. This resistance resulted from the induction of cIAP2 (encoded by BIRC3) since depletion of BIRC3 by siRNA or inhibition of cIAP2 via BV6 reversed H. pylori-suppressed caspase-3 activation. The induction of cIAP2 was regulated by H. pylori-induced BIRC3 eRNA synthesis. Depletion of BIRC3 eRNA decreased H. pylori-induced cIAP2 and reversed H. pylori-suppressed caspase-3 activation. Mechanistically, H. pylori stimulated the recruitment of bromodomain-containing factor Brd4 to the enhancer of BIRC3 and promoted BIRC3 eRNA and mRNA synthesis. Inhibition of Brd4 diminished the expression of BIRC3 eRNA and the anti-apoptotic response to H. pylori infection. Importantly, H. pylori isogenic cagA-deficient mutant failed to activate the synthesis of BIRC3 eRNA and the associated apoptosis resistance. Finally, in primary human gastric epithelial cells, H. pylori also induced resistance to Raptinal-triggered caspase-3 activation by activating the Brd4-dependent BIRC3 eRNA synthesis in a CagA-dependent manner. These results identify a novel function of Brd4 in H. pylori-mediated apoptosis resistance via activating BIRC3 eRNA synthesis, suggesting that Brd4 could be a potential therapeutic target for H. pylori-induced gastric cancer.
- Published
- 2020
5. Pinning Down the Transcription: A Role for Peptidyl-Prolyl cis-trans Isomerase Pin1 in Gene Expression
- Author
-
Xiangming Hu and Lin-Feng Chen
- Subjects
0301 basic medicine ,RNA polymerase II ,Isomerase ,isomerization ,03 medical and health sciences ,conformational change ,Pin1 ,0302 clinical medicine ,Transcription (biology) ,Gene expression ,lcsh:QH301-705.5 ,Gene ,Transcription factor ,biology ,phosphorylation ,Chemistry ,RNA ,Cell Biology ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,biology.protein ,PIN1 ,transcription ,Developmental Biology - Abstract
Pin1 is a peptidyl-prolyl cis-trans isomerase that specifically binds to a phosphorylated serine or threonine residue preceding a proline (pSer/Thr-Pro) motif and catalyzes the cis-trans isomerization of proline imidic peptide bond, resulting in conformational change of its substrates. Pin1 regulates many biological processes and is also involved in the development of human diseases, like cancer and neurological diseases. Many Pin1 substrates are transcription factors and transcription regulators, including RNA polymerase II (RNAPII) and factors associated with transcription initiation, elongation, termination and post-transcription mRNA decay. By changing the stability, subcellular localization, protein-protein or protein-DNA/RNA interactions of these transcription related proteins, Pin1 modulates the transcription of many genes related to cell proliferation, differentiation, apoptosis and immune response. Here, we will discuss how Pin regulates the properties of these transcription relevant factors for effective gene expression and how Pin1-mediated transcription contributes to the diverse pathophysiological functions of Pin1.
- Published
- 2020
6. <scp>PLK</scp> 4 deubiquitination by Spata2‐CYLD suppresses NEK7‐mediated NLRP3 inflammasome activation at the centrosome
- Author
-
Wenguo Li, Jun Qin, Shuangyan Zhang, Xiaoli Jiang, Qijun Wang, Xuefeng Wu, Zhiduo Liu, Dandan Wu, Rong Tan, Lin Feng Chen, Bing Su, Xiao Dong Yang, and Xiaoyin Niu
- Subjects
Inflammasomes ,Peritonitis ,Protein Serine-Threonine Kinases ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,Mice ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,NIMA-Related Kinases ,Centrosome duplication ,Phosphorylation ,Molecular Biology ,Centrosome ,Mice, Knockout ,Innate immune system ,integumentary system ,General Immunology and Microbiology ,Macrophages ,General Neuroscience ,Ubiquitination ,Proteins ,Inflammasome ,Articles ,Deubiquitinating Enzyme CYLD ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,Cytokines ,Inflammasome complex ,Signal Transduction ,medicine.drug ,Deubiquitination - Abstract
The innate immune sensor NLRP3 assembles an inflammasome complex with NEK7 and ASC to activate caspase‐1 and drive the maturation of proinflammatory cytokines IL‐1β and IL‐18. NLRP3 inflammasome activity must be tightly controlled, as its over‐activation is involved in the pathogenesis of inflammatory diseases. Here, we show that NLRP3 inflammasome activation is suppressed by a centrosomal protein Spata2. Spata2 deficiency enhances NLRP3 inflammasome activity both in the macrophages and in an animal model of peritonitis. Mechanistically, Spata2 recruits the deubiquitinase CYLD to the centrosome for deubiquitination of polo‐like kinase 4 (PLK4), the master regulator of centrosome duplication. Deubiquitination of PLK4 facilitates its binding to and phosphorylation of NEK7 at Ser204. NEK7 phosphorylation in turn attenuates NEK7 and NLRP3 interaction, which is required for NLRP3 inflammasome activation. Pharmacological or shRNA‐mediated inhibition of PLK4, or mutation of the NEK7 Ser204 phosphorylation site, augments NEK7 interaction with NLRP3 and causes increased NLRP3 inflammasome activation. Our study unravels a novel centrosomal regulatory pathway of inflammasome activation and may provide new therapeutic targets for the treatment of NLRP3‐associated inflammatory diseases.
- Published
- 2019
7. Multiple P-TEFbs cooperatively regulate the release of promoter-proximally paused RNA polymerase II
- Author
-
Xiaodong Lu, Min Liu, Ruichuan Chen, Meihua Chen, Songkuan Zhuang, Lin Feng Chen, Bin Yu, Haiyang Yang, Yanheng Chen, Runzhong Liu, Yao Wang, You Li, Muhua Rao, Kai Zhou, Yu Wang, and Xinxing Zhu
- Subjects
0301 basic medicine ,BRD4 ,Transcription Elongation, Genetic ,Cell Cycle Proteins ,RNA polymerase II ,Models, Biological ,MED1 ,03 medical and health sciences ,Mediator ,Transcription (biology) ,Acetamides ,Genetics ,Humans ,Positive Transcriptional Elongation Factor B ,Phosphorylation ,RNA, Small Interfering ,Promoter Regions, Genetic ,Molecular Biology ,biology ,Nuclear Proteins ,MED26 ,RNA ,HCT116 Cells ,DSIF ,Cell biology ,030104 developmental biology ,biology.protein ,RNA Polymerase II ,Transcriptional Elongation Factors ,HeLa Cells ,Transcription Factors - Abstract
The association of DSIF and NELF with initiated RNA Polymerase II (Pol II) is the general mechanism for inducing promoter-proximal pausing of Pol II. However, it remains largely unclear how the paused Pol II is released in response to stimulation. Here, we show that the release of the paused Pol II is cooperatively regulated by multiple P-TEFbs which are recruited by bromodomain-containing protein Brd4 and super elongation complex (SEC) via different recruitment mechanisms. Upon stimulation, Brd4 recruits P-TEFb to Spt5/DSIF via a recruitment pathway consisting of Med1, Med23 and Tat-SF1, whereas SEC recruits P-TEFb to NELF-A and NELF-E via Paf1c and Med26, respectively. P-TEFb-mediated phosphorylation of Spt5, NELF-A and NELF-E results in the dissociation of NELF from Pol II, thereby transiting transcription from pausing to elongation. Additionally, we demonstrate that P-TEFb-mediated Ser2 phosphorylation of Pol II is dispensable for pause release. Therefore, our studies reveal a co-regulatory mechanism of Brd4 and SEC in modulating the transcriptional pause release by recruiting multiple P-TEFbs via a Mediator- and Paf1c-coordinated recruitment network.
- Published
- 2016
8. T63 inhibits osteoclast differentiation through regulating MAPKs and Akt signaling pathways
- Author
-
Xiao li Zhao, Zhen Wang, Shu yi Si, Jin Jing Chen, and Lin Feng Chen
- Subjects
musculoskeletal diseases ,0301 basic medicine ,MAP Kinase Signaling System ,Osteoclasts ,Thiophenes ,Bone resorption ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Osteoclast ,medicine ,Animals ,Protein kinase B ,Pharmacology ,Osteoblasts ,biology ,Chemistry ,Kinase ,RANK Ligand ,Osteoblast ,Cell Differentiation ,Isoxazoles ,Actins ,Cell biology ,RUNX2 ,Enzyme Activation ,030104 developmental biology ,medicine.anatomical_structure ,RAW 264.7 Cells ,Gene Expression Regulation ,RANKL ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Proto-Oncogene Proteins c-akt - Abstract
Inhibition of excessive osteoclast differentiation and activity is a valid approach for the treatment of osteoporosis. T63 is a small-molecule compound identified from a high throughput screening based on RUNX2 transcriptional activity, and has been reported to stimulate osteoblast formation. However, whether the compound has any effect on osteoclast differentiation remains unknown. Here, we examined the in vitro effect of T63 on osteoclastogenesis. T63 was found to inhibit the number of TRAP-positive cells in an osteoblast-osteoclast co-culture system, and inhibited Rankl expression in the preosteoblast MC3T3-E1 cells. The compound also directly suppressed RANKL-induced osteoclast differentiation in both dose- and time-dependent manner, as evidenced by the decrease of TRAP activity, F-actin formation and osteoclastogenesis-related genes expression in RAW264.7 cells. Moreover, pretreatment with T63 markedly decreased the activation of mitogen-activated protein kinases and Akt, both of which are positively involved in the regulation of osteoclastogenesis. Collectively, our findings suggest T63 has a protective effect against bone loss by inhibiting bone resorption. Its regulatory effect on bone metabolism makes the compound a more promising candidate for the potential application in the treatment of osteoporosis.
- Published
- 2018
9. Small molecule T63 suppresses osteoporosis by modulating osteoblast differentiation via BMP and WNT signaling pathways
- Author
-
Guoning Zhang, Zhen Wang, Jin Jing Chen, Lin Feng Chen, Xiao li Zhao, Yucheng Wang, and Shu yi Si
- Subjects
0301 basic medicine ,medicine.medical_specialty ,lcsh:Medicine ,Biology ,Article ,Bone resorption ,Cell Line ,Bone remodeling ,Small Molecule Libraries ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,lcsh:Science ,Gene knockdown ,Osteoblasts ,Multidisciplinary ,lcsh:R ,Mesenchymal stem cell ,Wnt signaling pathway ,ALPL ,Cell Differentiation ,Osteoblast ,Alkaline Phosphatase ,High-Throughput Screening Assays ,Rats ,Cell biology ,Wnt Proteins ,RUNX2 ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Bone Morphogenetic Proteins ,Osteoporosis ,Female ,lcsh:Q ,Signal Transduction - Abstract
Osteoporosis results from the imbalance between bone resorption and bone formation, and restoring the normal balance of bone remodeling is highly desirable for identification of better treatment. In this study, using a cell-based high-throughput screening model representing Runt-related transcription factor 2 (RUNX2) transcriptional activity, we identified a novel small-molecular-weight compound, T63, as an efficient up-regulator of osteogenesis. T63 increased the alkaline phosphatase (ALPL) activity and mineralization as well as gene expression of Alpl and other osteogenic marker genes in mouse osteoblasts and mesenchymal stem cell-like cells. Upon induction of osteoblast differentiation, T63 inhibited adipogenic differentiation in the pluripotent mesenchymal cells. Consistently, T63 up-regulated RUNX2 mRNA and protein levels, and knockdown of RUNX2 reduced the osteogenic role of T63. Mechanistically, T63 activated both BMPs and WNT/β-catenin signaling pathways. Inhibition of either signaling pathway with specific inhibitor suppressed T63-induced RUNX2 expression and the osteogenic phenotypes. Moreover, T63 markedly protected against bone mass loss in the ovariectomized and dexamethasone treated rat osteoporosis model. Collectively, our data demonstrate that T63 could be a promising drug candidate and deserves further development for potential therapeutics in osteoporosis.
- Published
- 2017
10. Aesculin modulates bone metabolism by suppressing receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis and transduction signals
- Author
-
Xiao li Zhao, Lin Feng Chen, and Zhen Wang
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Biophysics ,Molecular Conformation ,Pharmacology ,Biochemistry ,Bone and Bones ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,0302 clinical medicine ,Osteoclast ,Osteogenesis ,Internal medicine ,medicine ,Cathepsin K ,Animals ,Molecular Biology ,Cells, Cultured ,Tartrate-resistant acid phosphatase ,biology ,Dose-Response Relationship, Drug ,Activator (genetics) ,RANK Ligand ,NF-κB ,Cell Biology ,Esculin ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,RAW 264.7 Cells ,chemistry ,RANKL ,030220 oncology & carcinogenesis ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
Aesculin (AES), a coumarin compound derived from Aesculus hippocasanum L, is reported to exert protective role against inflammatory diseases, gastric disease and cancer. However, direct effect of AES in bone metabolism is deficient. In this study, we examined the effects of AES on osteoclast (OC) differentiation in receptor activator of NF-κB ligand (RANKL)-induced RAW264.7 cells. AES inhibits the OC differentiation in both dose- and time-dependent manner within non-toxic concentrations, as analyzed by Tartrate Resistant Acid Phosphatase (TRAP) staining. The actin ring formation manifesting OC function is also decreased by AES. Moreover, expressions of osteoclastogenesis related genes Trap, Atp6v0d2, Cathepsin K and Mmp-9 are decreased upon AES treatment. Mechanistically, AES attenuates the activation of MAPKs and NF-κB activity upon RANKL induction, thus leading to the reduction of Nfatc1 mRNA expression. Moreover, AES inhibits Rank expression, and RANK overexpression markedly decreases AES's effect on OC differentiation and NF-κB activity. Consistently, AES protects against bone mass loss in the ovariectomized and dexamethasone treated rat osteoporosis model. Taken together, our data demonstrate that AES can modulate bone metabolism by suppressing osteoclastogenesis and related transduction signals. AES therefore could be a promising agent for the treatment of osteoporosis.
- Published
- 2017
11. Single-molecule sorting reveals how ubiquitylation affects substrate recognition and activities of FBH1 helicase
- Author
-
Lin Feng Chen, Maria Spies, Tokiha Masuda-Ozawa, Yeon Soo Seo, and Trish T. Hoang
- Subjects
DNA Replication ,Binding Sites ,biology ,DNA Helicases ,Ubiquitination ,Helicase ,DNA ,Genome Integrity, Repair and Replication ,RNA Helicase A ,Primosome ,Cell biology ,Protein Structure, Tertiary ,DNA-Binding Proteins ,HEK293 Cells ,Control of chromosome duplication ,Genetics ,biology.protein ,Replisome ,Humans ,Primase ,Rad51 Recombinase ,Replication protein A ,dnaB helicase - Abstract
DNA repair helicases function in the cell to separate DNA duplexes or remodel nucleoprotein complexes. These functions are influenced by sensing and signaling; the cellular pool of a DNA helicase may contain subpopulations of enzymes carrying different post-translational modifications and performing distinct biochemical functions. Here, we report a novel experimental strategy, single-molecule sorting, which overcomes difficulties associated with comprehensive analysis of heterologously modified pool of proteins. This methodology was applied to visualize human DNA helicase F-box– containing DNA helicase (FBH1) acting on the DNA structures resembling a stalled or collapsed replication fork and its interactions with RAD51 nucleoprotein filament. Individual helicase molecules isolated from human cells with their native post-translational modifications were analyzed using total internal reflection fluorescence microscopy. Separation of the activity trajectories originated from ubiquitylated and non-ubiquitylated FBH1 molecules revealed that ubiquitylation affects FBH1 interaction with the RAD51 nucleoprotein filament, but not its translocase and helicase activities.
- Published
- 2013
12. Posttranslational modifications of NF-κB: Another layer of regulation for NF-κB signaling pathway
- Author
-
Lin Feng Chen, Bo Huang, Xiao Dong Yang, and Acacia Lamb
- Subjects
Inflammation ,Programmed cell death ,Cell growth ,Eukaryotic transcription ,NF-kappa B ,Transcription Factor RelA ,NF-κB ,Cell Biology ,Biology ,NFKB1 ,Article ,Cell biology ,Mice ,chemistry.chemical_compound ,chemistry ,Cytoplasm ,Neoplasms ,Animals ,Humans ,Signal transduction ,Protein Processing, Post-Translational ,Gene ,Signal Transduction - Abstract
The eukaryotic transcription factor NF-kappaB regulates a wide range of host genes that control the inflammatory and immune responses, programmed cell death, cell proliferation and differentiation. The activation of NF-kappaB is tightly controlled both in the cytoplasm and in the nucleus. While the upstream cytoplasmic regulatory events for the activation of NF-kappaB are well studied, much less is known about the nuclear regulation of NF-kappaB. Emerging evidence suggests that NF-kappaB undergoes a variety of posttranslational modifications, and that these modifications play a key role in determining the duration and strength of NF-kappaB nuclear activity as well as its transcriptional output. Here we summarize the recent advances in our understanding of the posttranslational modifications of NF-kappaB, the interplay between the various modifications, and the physiological relevance of these modifications.
- Published
- 2010
13. Functional Interplay between Acetylation and Methylation of the RelA Subunit of NF-κB
- Author
-
Lin Feng Chen, Emad Tajkhorshid, and Xiao Dong Yang
- Subjects
Models, Molecular ,Glutamine ,Molecular Sequence Data ,Transcription Factor RelA ,Lysine ,Plasma protein binding ,Methylation ,Models, Biological ,complex mixtures ,Histone-Lysine N-Methyltransferase ,Cell Line ,Mice ,Ubiquitin ,Animals ,Humans ,Amino Acid Sequence ,Promoter Regions, Genetic ,Molecular Biology ,RELA ,biology ,Protein Stability ,Tumor Necrosis Factor-alpha ,Ubiquitination ,Acetylation ,Articles ,Cell Biology ,Protein Structure, Tertiary ,Protein Subunits ,Biochemistry ,Mutation ,biology.protein ,bacteria ,Protein Binding - Abstract
Posttranslational modifications of the RelA subunit of NF-kappaB, including acetylation and methylation, play a key role in controlling the strength and duration of its nuclear activity. Whether these modifications are functionally linked is largely unknown. Here, we show that the acetylation of lysine 310 of RelA impairs the Set9-mediated methylation of lysines 314 and 315, which is important for the ubiquitination and degradation of chromatin-associated RelA. Abolishing the acetylation of lysine 310 either by the deacetylase SIRT1 or by mutating lysine 310 to arginine enhances methylation. Conversely, enhancing the acetylation of lysine 310 by depleting SIRT1 or by replacing lysine 310 with acetyl-mimetic glutamine inhibits methylation, thereby decreasing ubiquitination, prolonging the stability of chromatin-associated RelA, and enhancing the transcriptional activity of NF-kappaB. The acetylation of lysine 310 of RelA interferes with its interaction with Set9. Based on structural modeling of the SET domain of Set9 with RelA, we propose that the positive charge of lysine 310 is critical for the binding of RelA to a negatively charged "exosite" within the SET domain of Set9. Together, these findings demonstrate for the first time an interplay between RelA acetylation and methylation and also provide a novel mechanism for the regulation of lysine methylation by acetylation.
- Published
- 2010
14. Human Immunodeficiency Virus Type 1 Tat Protein Inhibits the SIRT1 Deacetylase and Induces T Cell Hyperactivation
- Author
-
Ronen Marmorstein, Ruth Getachew, Warner C. Greene, Michael M. Brent, Michael W. McBurney, Melanie Ott, Prerana Jayakumar, Lin Feng Chen, Hye Sook Kwon, and Martina Schnölzer
- Subjects
Cancer Research ,MICROBIO ,T-Lymphocytes ,T cell ,Lymphocyte Activation ,Microbiology ,Article ,Cell Line ,Mice ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,Immune system ,Sirtuin 1 ,Immunology and Microbiology(all) ,Virology ,Protein Interaction Mapping ,medicine ,Animals ,Humans ,Sirtuins ,Molecular Biology ,Transcription factor ,030304 developmental biology ,0303 health sciences ,biology ,Hyperactivation ,Lysine ,Transcription Factor RelA ,3. Good health ,Cell biology ,medicine.anatomical_structure ,SIGNALING ,Cell culture ,030220 oncology & carcinogenesis ,HIV-1 ,biology.protein ,tat Gene Products, Human Immunodeficiency Virus ,Parasitology ,NAD+ kinase ,Protein Binding - Abstract
Symptoms of T cell hyperactivation shape the course and outcome of HIV-1 infection, but the mechanism(s) underlying this chronic immune activation are not well understood. We find that the viral transactivator Tat promotes hyperactivation of T cells by blocking the nicotinamide adenine dinucleotide (NAD(+))-dependent deacetylase SIRT1. Tat directly interacts with the deacetylase domain of SIRT1 and blocks the ability of SIRT1 to deacetylate lysine 310 in the p65 subunit of NF-kappaB. Because acetylated p65 is more active as a transcription factor, Tat hyperactivates the expression of NF-kappaB-responsive genes, a function lost in SIRT1-/- cells. These results support a model where the normal function of SIRT1 as a negative regulator of T cell activation is suppressed by Tat during HIV infection. These events likely contribute to the state of immune cell hyperactivation found in HIV-infected individuals.
- Published
- 2008
15. Synergistic activation of NF-κB by nontypeable H. influenzae and S. pneumoniae is mediated by CK2, IKKβ-IκBα, and p38 MAPK
- Author
-
Haidong Xu, Soo Mi Kweon, Lin Feng Chen, Hajime Ishinaga, Hirofumi Jono, Beinan Wang, Jae Hyang Lim, Jian Dong Li, Davida D. Rixter, and Tomoaki Koga
- Subjects
p38 mitogen-activated protein kinases ,Biophysics ,Inflammation ,NF-κB ,Cell Biology ,Biology ,medicine.disease_cause ,Biochemistry ,Microbiology ,chemistry.chemical_compound ,IκBα ,chemistry ,In vivo ,Streptococcus pneumoniae ,medicine ,medicine.symptom ,Casein kinase 2 ,Signal transduction ,Molecular Biology - Abstract
In review of the past studies on NF-kappaB regulation, most of them have focused on investigating how NF-kappaB is activated by a single inducer at a time. Given the fact that, in mixed bacterial infections in vivo, multiple inflammation inducers, including both nontypeable Haemophilus influenzae (NTHi) and Streptococcus pneumoniae, are present simultaneously, a key issue that has yet to be addressed is whether NTHi and S. pneumoniae simultaneously activate NF-kappaB and the subsequent inflammatory response in a synergistic manner. Here, we show that NTHi and S. pneumoniae synergistically induce NF-kappaB-dependent inflammatory response via activation of multiple signaling pathways in vitro and in vivo. The classical IKKbeta-IkappaBalpha and p38 MAPK pathways are involved in synergistic activation of NF-kappaB via two distinct mechanisms, p65 nuclear translocation-dependent and -independent mechanisms. Moreover, casein kinase 2 (CK2) is involved in synergistic induction of NF-kappaB via a mechanism dependent on phosphorylation of p65 at both Ser536 and Ser276 sites. These studies bring new insights into the molecular mechanisms underlying the NF-kappaB-dependent inflammatory response in polymicrobial infections and may lead to development of novel therapeutic strategies for modulating inflammation in mixed infections for patients with otitis media and chronic obstructive pulmonary diseases.
- Published
- 2006
16. NF-κB RelA Phosphorylation Regulates RelA Acetylation
- Author
-
James M. Duerr, Leonard Buckbinder, Hiroyasu Nakano, Yajun Mu, Warner C. Greene, Samuel A. Williams, and Lin Feng Chen
- Subjects
Transcription, Genetic ,Lysine ,Transcription Factor RelA ,Gene Expression ,Biology ,Serine ,Mice ,Animals ,Humans ,Phosphorylation ,Protein kinase A ,Molecular Biology ,Cells, Cultured ,RELA ,Kinase ,NF-kappa B ,Nuclear Proteins ,Acetylation ,Cell Biology ,Biochemistry ,Mutation ,Trans-Activators ,bacteria ,E1A-Associated p300 Protein - Abstract
The nuclear functions of NF-kappaB p50/RelA heterodimers are regulated in part by posttranslational modifications of its RelA subunit, including phosphorylation and acetylation. Acetylation at lysines 218, 221, and 310 differentially regulates RelA's DNA binding activity, assembly with IkappaBalpha, and transcriptional activity. However, it remains unclear whether the acetylation is regulated or simply due to stimulus-coupled nuclear translocation of NF-kappaB. Using anti-acetylated lysine 310 RelA antibodies, we detected p300-mediated acetylation of RelA in vitro and in vivo after stimulation of cells with tumor necrosis factor alpha (TNF-alpha). Coexpression of catalytically inactive mutants of the catalytic subunit of protein kinase A/mitogen- and stress-activated kinase 1 or IKK1/IKK2, which phosphorylate RelA on serine 276 or serine 536, respectively, sharply inhibited RelA acetylation on lysine 310. Furthermore, phosphorylation of RelA on serine 276 or serine 536 increased assembly of phospho-RelA with p300, which enhanced acetylation on lysine 310. Reconstitution of RelA-deficient murine embryonic fibroblasts with RelA S276A or RelA S536A decreased TNF-alpha-induced acetylation of lysine 310 and expression of the endogenous NF-kappaB-responsive E-selectin gene. These findings indicate that the acetylation of RelA at lysine 310 is importantly regulated by prior phosphorylation of serines 276 and 536. Such phosphorylated and acetylated forms of RelA display enhanced transcriptional activity.
- Published
- 2005
17. NF-κB Is Essential for Induction of CYLD, the Negative Regulator of NF-κB
- Author
-
Eirini Trompouki, George Mosialos, Hirofumi Jono, Jian Dong Li, Zhixing K. Pan, Lin Feng Chen, Haidong Xu, and Jae Hyang Lim
- Subjects
Regulation of gene expression ,TRAF2 ,I-Kappa-B Kinase ,NF-κB ,Cell Biology ,Biology ,medicine.disease_cause ,Biochemistry ,Deubiquitinating Enzyme CYLD ,Cell biology ,chemistry.chemical_compound ,chemistry ,Immunology ,medicine ,Tumor necrosis factor alpha ,Carcinogenesis ,Molecular Biology ,Transcription factor - Abstract
The transcription factor NF-kappaB regulates genes involved in inflammatory and immune responses, tumorigenesis, and apoptosis. In contrast to the pleiotropic stimuli that lead to its positive regulation, the known signaling mechanisms that underlie the negative regulation of NF-kappaB are very few. Recent studies have identified the tumor suppressor CYLD, loss of which causes a benign human syndrome called cylindromatosis, as a key negative regulator for NF-kappaB signaling by deubiquitinating tumor necrosis factor (TNF) receptor-associated factor (TRAF) 2, TRAF6, and NEMO (NF-kappaB essential modulator, also known as IkappaB kinase gamma). However, how CYLD is regulated remains unknown. The present study revealed a novel autoregulatory feedback pathway through which activation of NF-kappaB by TNF-alpha and bacterium nontypeable Haemophilus influenzae (NTHi) induces CYLD that in turn leads to the negative regulation of NF-kappaB signaling. In addition, TRAF2 and TRAF6 appear to be differentially involved in NF-kappaB-dependent induction of CYLD by TNF-alpha and NTHi. These findings provide novel insights into the autoregulation of NF-kappaB activation.
- Published
- 2004
18. p53 Induces NF-κB Activation by an IκB Kinase-independent Mechanism Involving Phosphorylation of p65 by Ribosomal S6 Kinase 1
- Author
-
Yajun Mu, Jan Bohuslav, Hakju Kwon, Lin Feng Chen, and Warner C. Greene
- Subjects
Cytoplasm ,Transcription, Genetic ,Apoptosis ,Protein Serine-Threonine Kinases ,Biology ,Mitogen-activated protein kinase kinase ,Transfection ,Models, Biological ,Ribosomal Protein S6 Kinases, 90-kDa ,Biochemistry ,Cell Line ,MAP2K7 ,Mice ,TANK-binding kinase 1 ,Genes, Reporter ,Animals ,Humans ,ASK1 ,Gene Silencing ,Phosphorylation ,RNA, Small Interfering ,Molecular Biology ,Cell Nucleus ,Serine/threonine-specific protein kinase ,MAP kinase kinase kinase ,NF-kappa B ,Transcription Factor RelA ,I-Kappa-B Kinase ,Cell Biology ,Fibroblasts ,Precipitin Tests ,I-kappa B Kinase ,Enzyme Activation ,Mutation ,Cyclin-dependent kinase 9 ,Tumor Suppressor Protein p53 ,DNA Damage ,Protein Binding - Abstract
Apoptosis induced by p53 has been proposed to involve activation of the transcription factor NF-kappaB. Here we describe the novel molecular mechanism through which p53 and DNA-damaging agents activate NF-kappaB. NF-kappaB induction by p53 does not occur through classical activation of the IkappaB kinases and degradation of IkappaBalpha. Rather, p53 expression stimulates the serine/threonine kinase ribosomal S6 kinase 1 (RSK1), which in turn phosphorylates the p65 subunit of NF-kappaB. The lower affinity of RSK1-phosphorylated p65 for its negative regulator, IkappaBalpha, decreases IkappaBalpha-mediated nuclear export of shuttling forms of NF-kappaB, thereby promoting the binding and action of NF-kappaB on cognate kappaB enhancers. These findings highlight a rather unusual pathway of NF-kappaB activation, which is utilized by the p53 tumor suppressor.
- Published
- 2004
19. Shaping the nuclear action of NF-κB
- Author
-
Warner C. Greene and Lin Feng Chen
- Subjects
biology ,Cell growth ,NF-κB ,Cell Biology ,Cell biology ,Cell nucleus ,chemistry.chemical_compound ,Histone ,medicine.anatomical_structure ,Immune system ,chemistry ,biology.protein ,medicine ,Phosphorylation ,Molecular Biology ,Gene ,Transcription factor - Abstract
The NF-κB/REL family of transcription factors pivotally control the inflammatory and immune responses, as well as other genetic programmes that are central to cell growth and survival. The cytoplasmic regulation of NF-κB is well characterized and, recently, significant progress has been made in understanding how its nuclear action is regulated. Post-translational modification of the NF-κB subunits as well as histones surrounding the NF-κB target genes has a key role in this regulation. Here, we review the important advances that constitute this new and exciting chapter in NF-κB biology.
- Published
- 2004
20. Talking to histone: methylated RelA serves as a messenger
- Author
-
Lin Feng Chen and Xiao Dong Yang
- Subjects
Cellular differentiation ,Transcription Factor RelA ,Regulator ,Apoptosis ,IκB kinase ,Methylation ,Histones ,Animals ,Humans ,Molecular Biology ,Transcription factor ,Cell Proliferation ,Inflammation ,biology ,NF-kappa B ,I-Kappa-B Kinase ,Cell Differentiation ,Cell Biology ,Acquired immune system ,Research Highlight ,I-kappa B Kinase ,Cell biology ,Histone ,biology.protein ,Cancer research - Abstract
The mammalian NF-κB family of transcription factors functions as a major regulator of innate and adaptive immunity and inflammatory responses, plays critical roles in controlling cell proliferation, differentiation and apoptosis by regulating a wide range of genes that govern these various processes 1. The prototypical NF-κB (p50 and RelA heterodimer) is sequestered and inactivated in the cytoplasm by a family of inhibitors, called IκBs, in unstimulated cells. NF-κB is activated in response to a variety of stimuli, including inflammatory cytokines and viral and bacterial infections, which lead to the activation of the IκB kinase complex (IKK), phosphorylation and degradation of IκBs, and the nuclear translocation of the NF-κB heterodimer.
- Published
- 2011
21. Interaction and Functional Cooperation of PEBP2/CBF with Smads
- Author
-
Wei-hui Guo, Y Ishidou, Masahiro Kawabata, Naoko Ohtani-Fujita, Minoru Fukuchi, Janet Stavnezer, Lin Feng Chen, Yoshiaki Ito, Meng Jiao Shi, Takeshi Imamura, Kohei Miyazono, Tomohiko Kanno, Jun-ichi Hanai, and Woo-Young Kim
- Subjects
Core Binding Factor alpha Subunits ,Transcription factor complex ,Cell Biology ,Transforming growth factor beta ,SMAD ,Biology ,Core binding factor ,Biochemistry ,Molecular biology ,Sp3 transcription factor ,Enhancer binding ,biology.protein ,Molecular Biology ,Transcription factor - Abstract
Smads are signal transducers for members of the transforming growth factor-beta (TGF-beta) superfamily. Upon ligand stimulation, receptor-regulated Smads (R-Smads) are phosphorylated by serine/threonine kinase receptors, form complexes with common-partner Smad, and translocate into the nucleus, where they regulate the transcription of target genes together with other transcription factors. Polyomavirus enhancer binding protein 2/core binding factor (PEBP2/CBF) is a transcription factor complex composed of alpha and beta subunits. The alpha subunits of PEBP2/CBF, which contain the highly conserved Runt domain, play essential roles in hematopoiesis and osteogenesis. Here we show that three mammalian alpha subunits of PEBP2/CBF form complexes with R-Smads that act in TGF-beta/activin pathways as well as those acting in bone morphogenetic protein (BMP) pathways. Among them, PEBP2alphaC/CBFA3/AML2 forms a complex with Smad3 and stimulates transcription of the germline Ig Calpha promoter in a cooperative manner, for which binding of both factors to their specific binding sites is essential. PEBP2 may thus be a nuclear target of TGF-beta/BMP signaling.
- Published
- 1999
22. Bromodomain and Extraterminal (BET) Protein Inhibition Suppresses Human T Cell Leukemia Virus 1 (HTLV-1) Tax Protein-mediated Tumorigenesis by Inhibiting Nuclear Factor κB (NF-κB) Signaling*
- Author
-
Lin Feng Chen, Xuewei Wu, Jun Qi, Gutian Xiao, and James E. Bradner
- Subjects
BRD4 ,Carcinogenesis ,Transcription Factor RelA ,T-cell leukemia ,Down-Regulation ,Cell Cycle Proteins ,Biology ,Biochemistry ,Cell Line ,Mice ,Animals ,Humans ,Nuclear protein ,Molecular Biology ,Transcription factor ,Cell Proliferation ,Cell Nucleus ,Human T-lymphotropic virus 1 ,RELA ,Lysine ,Nuclear Proteins ,Acetylation ,Cell Biology ,Azepines ,Gene Products, tax ,Triazoles ,Bromodomain ,Gene Expression Regulation, Neoplastic ,Cancer research ,Signal transduction ,Signal Transduction ,Transcription Factors - Abstract
The etiology of human T cell leukemia virus 1 (HTLV-1)-mediated adult T cell leukemia is associated with the ability of viral oncoprotein Tax to induce sustained NF-κB activation and the expression of many NF-κB target genes. Acetylation of the RelA subunit of NF-κB and the subsequent recruitment of bromodomain-containing factor Brd4 are important for the expression of NF-κB target genes in response to various stimuli. However, their contributions to Tax-mediated NF-κB target gene expression and tumorigenesis remain unclear. Here we report that Tax induced the acetylation of lysine 310 of RelA and the binding of Brd4 to acetylated RelA to facilitate Tax-mediated transcriptional activation of NF-κB. Depletion of Brd4 down-regulated Tax-mediated NF-κB target gene expression and cell proliferation. Inhibiting the interaction of Brd4 and acetylated RelA with the bromodomain extraterminal protein inhibitor JQ1 suppressed the proliferation of Tax-expressing rat fibroblasts and Tax-positive HTLV-1-infected cells and Tax-mediated cell transformation and tumorigenesis. Moreover, JQ1 attenuated the Tax-mediated transcriptional activation of NF-κB, triggering the polyubiquitination and proteasome-mediated degradation of constitutively active nuclear RelA. Our results identify Brd4 as a key regulator for Tax-mediated NF-κB gene expression and suggest that targeting epigenetic regulators such as Brd4 with the bromodomain extraterminal protein inhibitor might be a potential therapeutic strategy for cancers and other diseases associated with HTLV-1 infection.
- Published
- 2013
23. Role of the Helicobacter pylori-induced inflammatory response in the development of gastric cancer
- Author
-
Acacia Lamb and Lin Feng Chen
- Subjects
Inflammation ,medicine.disease_cause ,Biochemistry ,Article ,Helicobacter Infections ,Bacterial Proteins ,Stomach Neoplasms ,medicine ,Humans ,Molecular Biology ,Antigens, Bacterial ,biology ,Helicobacter pylori ,NF-kappa B ,Cancer ,Cell Biology ,Hepatitis B ,biology.organism_classification ,medicine.disease ,MAP Kinase Kinase Kinases ,Enzyme Activation ,Leukemia ,Chronic infection ,Gastric Mucosa ,Gastritis ,Immunology ,Host-Pathogen Interactions ,medicine.symptom ,Carcinogenesis - Abstract
Many factors are known to contribute to the development of cancer, and gastric cancer development is no different. Chronic infections, tobacco smoke, diet, and obesity are all major factors which play into the development of cancer [Aggarwal and Gehlot, 2009]. There is a single mechanism which underlies many of these risk factors: inflammation. The link between inflammation and cancer was proposed by Virchow in the 19th century with his observation that tumors often arose at sites of chronic inflammation and that inflammatory cells were present in tumor samples [Balkwill and Mantovani, 2001]. Inflammation has been observed in many infection-triggered cancers, and approximately 15% of human cancers are associated with chronic infection and inflammation [Karin and Greten, 2005]. For example, Hepatitis B or C virus infections, given their names because of their ability to cause chronic liver inflammation, can lead to hepatocellular carcinoma. Infections with many types of human papillomaviruses are recognized as key risk factors for cervical cancer, and the inflammation caused by persistent infection acts as a “cofactor” in carcinogenesis. A protein product of human T-cell leukemia/lymphotrophic virus type 1 (HTLV-1) “hijacks” host inflammation-regulating pathways to induce inflammation and transform T-cells, leading to the development of adult T-cell leukemia [Sun and Yamaoka, 2005]. Inflammation has become a new hallmark for cancer [Hanahan and Weinberg, 2011].
- Published
- 2013
24. New insights into the inactivation of gastric tumor suppressor RUNX3: the role of H. pylori infection
- Author
-
Acacia Lamb, Lin Feng Chen, and Ying Hung Nicole Tsang
- Subjects
Biochemistry ,Article ,law.invention ,Helicobacter Infections ,Pathogenesis ,law ,Stomach Neoplasms ,medicine ,Animals ,Humans ,Risk factor ,Molecular Biology ,Transcription factor ,biology ,Helicobacter pylori ,digestive, oral, and skin physiology ,Cancer ,Cell Biology ,biology.organism_classification ,H pylori infection ,medicine.disease ,digestive system diseases ,Core Binding Factor Alpha 3 Subunit ,Immunology ,DNA methylation ,Suppressor - Abstract
Runt-related transcription factor 3, or RUNX3, is a tumor suppressor in gastric cancer. Inactivation of RUNX3 is causally associated with the genesis of gastric cancer, since RUNX3 is frequently inactivated in gastric cancers by hemizygous deletion, hypermethylation of its promoter, or protein mislocalization. Infection with Helicobacter pylori is the strongest risk factor for the development of gastric cancer. Recent studies have indicated that H. pylori infection plays an important role in the inactivation of RUNX3, and that this inactivation contributes to the pathogenesis of H. pylori. Here we summarize these recent advances and discuss their significances in understanding the initiation and development of gastric cancer.
- Published
- 2011
25. Negative regulation of NF-κB action by Set9-mediated lysine methylation of the RelA subunit
- Author
-
Mingxi Li, Acacia Lamb, Lin Feng Chen, Neil L. Kelleher, Xiao Dong Yang, and Bo Huang
- Subjects
Transcriptional Activation ,Methyltransferase ,Recombinant Fusion Proteins ,Transcription Factor RelA ,Molecular Sequence Data ,Biology ,Histone-Lysine N-Methyltransferase ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,chemistry.chemical_compound ,RNA interference ,Humans ,Amino Acid Sequence ,Protein Methyltransferases ,Promoter Regions, Genetic ,Molecular Biology ,RELA ,General Immunology and Microbiology ,Tumor Necrosis Factor-alpha ,General Neuroscience ,Lysine ,NF-kappa B ,NF-κB ,Molecular biology ,Cell biology ,Protein Subunits ,chemistry ,Histone methyltransferase ,Histone Methyltransferases ,RNA Interference - Abstract
Proper regulation of NF-kappaB activity is critical to maintain and balance the inflammatory response. Inactivation of the NF-kappaB complex relies in part on the proteasome-mediated degradation of promoter-bound NF-kappaB, but the detailed molecular mechanism initiating this process remains elusive. Here, we show that the methylation of the RelA subunit of NF-kappaB has an important function in this process. Lysine methyltransferase Set9 physically associates with RelA in vitro and in vivo in response to TNF-alpha stimulation. Mutational and mass spectrometric analyses reveal that RelA is monomethylated by Set9 at lysine residues 314 and 315 in vitro and in vivo. Methylation of RelA inhibits NF-kappaB action by inducing the proteasome-mediated degradation of promoter-associated RelA. Depletion of Set9 by siRNA or mutation of the RelA methylation sites prolongs DNA binding of NF-kappaB and enhances TNF-alpha-induced expression of NF-kappaB target genes. Together, these findings unveil a novel mechanism by which methylation of RelA dictates the turnover of NF-kappaB and controls the NF-kappaB-mediated inflammatory response.
- Published
- 2009
26. Brd4 Coactivates Transcriptional Activation of NF-κB via Specific Binding to Acetylated RelA ▿
- Author
-
Xiao Dong Yang, Keiko Ozato, Ming Ming Zhou, Bo Huang, and Lin Feng Chen
- Subjects
Transcriptional Activation ,BRD4 ,RNA polymerase II ,Cell Cycle Proteins ,complex mixtures ,Cell Line ,Mice ,Transcription (biology) ,Coactivator ,Animals ,Humans ,Binding site ,Molecular Biology ,RELA ,Binding Sites ,biology ,Lysine ,NF-kappa B ,Transcription Factor RelA ,Nuclear Proteins ,Acetylation ,Cell Biology ,Articles ,DNA Polymerase II ,Molecular biology ,Cyclin-Dependent Kinase 9 ,Cell biology ,Bromodomain ,biology.protein ,bacteria ,Protein Binding ,Transcription Factors - Abstract
Acetylation of the RelA subunit of NF-kappaB, especially at lysine-310, is critical for the transcriptional activation of NF-kappaB and the expression of inflammatory genes. In this study, we demonstrate that bromodomains of Brd4 bind to acetylated lysine-310. Brd4 enhances transcriptional activation of NF-kappaB and the expression of a subset of NF-kappaB-responsive inflammatory genes in an acetylated lysine-310-dependent manner. Bromodomains of Brd4 and acetylated lysine-310 of RelA are both required for the mutual interaction and coactivation function of Brd4. Finally, we demonstrate that Brd4 further recruits CDK9 to phosphorylate C-terminal domain of RNA polymerase II and facilitate the transcription of NF-kappaB-dependent inflammatory genes. Our results identify Brd4 as a novel coactivator of NF-kappaB through specifically binding to acetylated lysine-310 of RelA. In addition, these studies reveal a mechanism by which acetylated RelA stimulates the transcriptional activity of NF-kappaB and the NF-kappaB-dependent inflammatory response.
- Published
- 2008
27. Regulation of NF-κB Action by Reversible Acetylation
- Author
-
Lin Feng Chen and Warner C. Greene
- Subjects
chemistry.chemical_compound ,RELA ,chemistry ,Biochemistry ,Acetylation ,Protein subunit ,Lysine ,Biology ,Nuclear export signal ,Subcellular localization ,HDAC3 ,DNA ,Cell biology - Abstract
While the proximal cytoplasmic signalling events controlling the activation of NF-kappaB are understood in considerable detail, the subsequent intranuclear events that regulate the strength and duration of NF-kappaB action remain poorly defined. Recently, we have demonstrated that the RelA subunit of the NF-kappaB heterodimer is subject to reversible acetylation. The p300/CBP acetyltransferases play a major role in the in vivo acetylation of RelA principally targeting lysines 218, 221 and 310 for modification. Acetylation of these distinct lysine residues regulates different functions of NF-kappaB, including transcriptional activation, DNA binding affinity, I-kappaBalpha assembly and subcellular localization. Specifically, acetylation of lysine 221 enhances DNA binding and impairs assembly with I-kappaBalpha while acetylation of lysine 310 is required for full transcriptional activity of RelA independent of changes in DNA binding or I-kappaBalpha binding. In turn, acetylated RelA is deacetylated by histone deacetylase 3 (HDAC3). Deacetylation of lysine 221 promotes high-affinity binding of RelA to newly synthesized I-kappaBalpha proteins whose expression is activated by NF-kappaB. I-kappaBalpha binding to deacetylated RelA promotes rapid nuclear export of the NF-kappaB complex. This export is dependent on CRM1 binding to a nuclear export signal present in I-KBalpha and promotes replenishment of the cytoplasmic pool of latent NF-kappaB/I-kappaBalpha complexes thus readying the cell for response to the next NF-kappaB inducing stimulus Together, these studies highlight how reversible acetylation of RelA serves as an intranuclear molecular switch promoting both positive and negative regulatory effects on nuclear NF-kappaB action.
- Published
- 2008
28. Synergistic induction of nuclear factor-kappaB by transforming growth factor-beta and tumour necrosis factor-alpha is mediated by protein kinase A-dependent RelA acetylation
- Author
-
Lin Feng Chen, Chen Yan, Jiyun Lee, Jian Dong Li, Xiangbin Xu, Kensei Komatsu, Xin-Hua Feng, Chang Hoon Woo, Hirofumi Jono, Hajime Ishinaga, Haidong Xu, and Jae Hyang Lim
- Subjects
TGF alpha ,MAP kinase kinase kinase ,Akt/PKB signaling pathway ,Tumor Necrosis Factor-alpha ,NF-kappa B ,Transcription Factor RelA ,Acetylation ,Cell Biology ,DNA ,Biology ,Mitogen-activated protein kinase kinase ,Biochemistry ,Cyclic AMP-Dependent Protein Kinases ,Proinflammatory cytokine ,Transforming Growth Factor beta ,Cell Line, Tumor ,Cancer research ,Humans ,ASK1 ,Protein kinase A ,Molecular Biology ,Transforming growth factor ,Protein Binding - Abstract
The TGF-beta (transforming growth factor-beta) pathway represents an important signalling pathway involved in regulating diverse biological processes, including cell proliferation, differentiation and inflammation. Despite the critical role for TGF-beta in inflammatory responses, its role in regulating NF-kappaB (nuclear factor-kappaB)-dependent inflammatory responses still remains unknown. In the present study we show that TGF-beta1 synergizes with proinflammatory cytokine TNF-alpha (tumour necrosis factor-alpha) to induce NF-kappaB activation and the resultant inflammatory response in vitro and in vivo. TGF-beta1 synergistically enhances TNF-alpha-induced NF-kappaB DNA binding activity via induction of RelA acetylation. Moreover, synergistic enhancement of TNF-alpha-induced RelA acetylation and DNA-binding activity by TGF-beta1 is mediated by PKA (protein kinase A). Thus the present study reveals a novel role for TGF-beta in inflammatory responses and provides new insight into the regulation of NF-kappaB by TGF-beta signalling.
- Published
- 2008
29. Regulation of Nuclear NF-_B Action
- Author
-
Lin-feng Chen and Warner Greene
- Subjects
Action (philosophy) ,Chemistry ,Key (cryptography) ,Posttranslational modification ,Cell biology - Published
- 2006
30. SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling
- Author
-
Saili Yi, Li Gan, Lin Feng Chen, Yungui Zhou, Lennart Mucke, Jennifer Y. Chen, Sarah Mueller-Steiner, and Hakju Kwon
- Subjects
Genetic Vectors ,Green Fluorescent Proteins ,Resveratrol ,Biochemistry ,Neuroprotection ,Models, Biological ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Sirtuin 1 ,Alzheimer Disease ,Genes, Reporter ,Stilbenes ,medicine ,Animals ,Humans ,Sirtuins ,Molecular Biology ,Cells, Cultured ,Neurons ,Amyloid beta-Peptides ,Microglia ,biology ,Models, Genetic ,Reverse Transcriptase Polymerase Chain Reaction ,Lysine ,Neurodegeneration ,Lentivirus ,Neurotoxicity ,NF-kappa B ,Cell Biology ,medicine.disease ,Immunohistochemistry ,Cell biology ,Rats ,IκBα ,medicine.anatomical_structure ,chemistry ,Bromodeoxyuridine ,Microscopy, Fluorescence ,Immunology ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
Accumulating evidence suggests that neurodegeneration induced by pathogenic proteins depends on contributions from surrounding glia. Here we demonstrate that NF-kappaB signaling in microglia is critically involved in neuronal death induced by amyloid-beta (Abeta) peptides, which are widely presumed to cause Alzheimer disease. Constitutive inhibition of NF-kappaB signaling in microglia by expression of the nondegradable IkappaBalpha superrepressor blocked neurotoxicity, indicating a pivotal role for microglial NF-kappaB signaling in mediating Abeta toxicity. Stimulation of microglia with Abeta increased acetylation of RelA/p65 at lysine 310, which regulates the NF-kappaB pathway. Overexpression of SIRT1 deacetylase and the addition of the SIRT1 agonist resveratrol markedly reduced NF-kappaB signaling stimulated by Abeta and had strong neuroprotective effects. Our results support a glial loop hypothesis by demonstrating a critical role for microglial NF-kappaB signaling in Abeta-dependent neurodegeneration. They also implicate SIRT1 in this pathway and highlight the therapeutic potential of resveratrol and other sirtuin-activating compounds in Alzheimer disease.
- Published
- 2005
31. Prostratin antagonizes HIV latency by activating NF-kappaB
- Author
-
Dwayne Bisgrove, Warner C. Greene, Hakju Kwon, David Fenard, Lin Feng Chen, Samuel A. Williams, and Eric Verdin
- Subjects
CD4-Positive T-Lymphocytes ,Gene Expression Regulation, Viral ,T cell ,Active Transport, Cell Nucleus ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Jurkat cells ,chemistry.chemical_compound ,Jurkat Cells ,Phorbol Esters ,medicine ,Humans ,Phosphorylation ,Prostratin ,Molecular Biology ,Transcription factor ,Protein kinase C ,Protein Kinase C ,HIV Long Terminal Repeat ,NF-kappa B ,HIV ,NFAT ,Cell Biology ,Virology ,Cell biology ,I-kappa B Kinase ,Virus Latency ,IκBα ,medicine.anatomical_structure ,chemistry ,Chromatin immunoprecipitation - Abstract
A subset of quiescent memory CD4 T cells harboring integrated but transcriptionally silent proviruses poses a currently insurmountable barrier to the eradication of the human immunodeficiency virus (HIV) in infected patients. Induction of HIV gene expression in these latently infected cells by immune activating agents has been proposed as one approach to confer sensitivity to antiretroviral therapy. Interest has recently focused on the non-tumor-promoting phorbol ester, prostratin, as a potential agent to activate latent HIV proviruses. Using multiple Jurkat T cell lines containing integrated but transcriptionally latent HIV proviruses (J-Lat cells), we now demonstrate that prostratin effectively activates HIV gene expression in these latently infected cells. We further show that prostratin acts by stimulating IKK-dependent phosphorylation and degradation of IkappaBalpha, leading to the rapid nuclear translocation of NF-kappaB and activation of the HIV-1 long terminal repeat in a kappaB enhancer-dependent manner. In contrast, NFAT and AP-1 are not induced by prostratin. Using chromatin immunoprecipitation assays to identify host transcription factors recruited to the latent HIV-1 promoter in living cells, we find that prostratin induces RelA binding. Analysis of potential upstream signal transducers demonstrates that prostratin stimulates membrane translocation of classical, novel, and atypical protein kinase C (PKC) isoforms. Studies with isoform-specific PKC inhibitors suggest that the novel PKCs play a particularly prominent role in the prostratin response. These findings provide new insights into the molecular pathway through which prostratin antagonizes HIV latency highlighting a central role for the action of NF-kappaB.
- Published
- 2004
32. Human T-cell lymphotropic virus type 1 tax induction of biologically Active NF-kappaB requires IkappaB kinase-1-mediated phosphorylation of RelA/p65
- Author
-
Lin Feng Chen, Alison O'Mahony, Katrien Van Beneden, Warner C. Greene, and Mauricio Montano
- Subjects
Transcriptional Activation ,Transcription Factor RelA ,IκB kinase ,Biology ,Protein Serine-Threonine Kinases ,Transfection ,Biochemistry ,Transactivation ,Mice ,Tumor Cells, Cultured ,Animals ,Humans ,Human T cell lymphotropic virus type 1 ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Kinase ,I-Kappa-B Kinase ,NF-kappa B ,Cell Biology ,Gene Products, tax ,Cell Transformation, Viral ,Cell biology ,I-kappa B Kinase ,Enzyme Activation ,Cancer research ,Signal transduction - Abstract
Activation of the NF-kappaB/Rel family of transcription factors proceeds through a catalytic complex containing IkappaB kinase (IKK)-1 and IKK2. Targeted disruption of each of the IKK genes suggests that these two kinases may mediate distinct functions in the activation pathway. In our studies of the human T-cell lymphotropic virus type 1 (HTLV-1) Tax oncoprotein, we have uncovered a new function of IKK1 required for complete activation of the NF-kappaB transcriptional program. In IKK1(-/-) murine embryonic fibroblasts (MEFs), Tax normally induced early NF-kappaB activation events. However, NF-kappaB induced by Tax in these IKK1(-/-) cells was functionally impaired. In IKK1(-/-) (but not wild-type) MEFs, Tax failed to activate several different kappaB reporter constructs or to induce the endogenous IkappaBalpha gene. In contrast, Tax normally activated the cAMP-responsive element-binding protein/activating transcription factor pathway, leading to full stimulation of an HTLV-1 long terminal repeat reporter construct in IKK1(-/-) cells. Furthermore, reconstitution of IKK1(-/-) cells with kinase-proficient (but not kinase-deficient) forms of IKK1 restored the Tax induction of full NF-kappaB transactivation. We further found that the defect in NF-kappaB action in IKK1(-/-) cells correlated with a failure of Tax to induce phosphorylation of the RelA/p65 subunit of NF-kappaB at Ser(529) and Ser(536). Such phosphorylation of RelA/p65 was readily detected in wild-type MEFs. Phosphorylation of Ser(536) was required for a complete response to Tax expression, whereas phosphorylation of Ser(529) appeared to be less critical. Together, these findings highlight distinct roles for the IKK1 and IKK2 kinases in the activation of NF-kappaB in response to HTLV-1 Tax. IKK2 plays a dominant role in signaling for IkappaBalpha degradation, whereas IKK1 appears to play an important role in enhancing the transcriptional activity of NF-kappaB by promoting RelA/p65 phosphorylation.
- Published
- 2004
33. New Insights into the Regulation of Nf-кB
- Author
-
W. C. Greene, A. O’Mahony, and Lin Feng Chen
- Subjects
Rel homology domain ,medicine.anatomical_structure ,Acetylation ,Cytoplasm ,Chemistry ,medicine ,Phosphorylation ,Casein kinase 1 ,Nuclear export signal ,Transcription factor ,Nucleus ,Cell biology - Abstract
Long considered to be a pivotal transcription factor regulating both the immune and inflammatory responses, NF-кB is now known to regulate such diverse processes as cell proliferation, differentiation and apoptotic cell death. NF-кB belongs to a family of transcription factors, all sharing an N-terminal Rel homology domain. NF-кB is tightly regulated within the cell at multiple levels, both during its activation and mobilization from the cytoplasm and before or during its transcriptional activation in the nucleus. Considering the wide variety of signals activating this transcription factor and the relative transcriptional strength of the RelA subunit, it is not surprising that NF-кB is regulated at multiple levels. The classical pathway leading to NF-кB activation proceeds via a kinase cascade, culminating in phosphorylation and degradation of IкBα, one member of a family of inhibitors controlling NF-кB action (Senftleben & Karin, 2002). NF-кB activation can be viewed as two discrete phases. The first comprises the proximal events leading to degradation of IкBα and rapid translocation of the liberated NF-кB complex into the nucleus. The second phase involves posttranslational modification of the NF-кB subunits themselves by both phosphorylation and acetylation, which are required for full activity of the induced nuclear NF-кB complex. While acetylation appears to occur in the nucleus, certain phosphorylation events in the second phase are likely executed in the cytoplasm before nuclear entry of NF-кB.
- Published
- 2003
34. Acetylation of RelA at discrete sites regulates distinct nuclear functions of NF-kappaB
- Author
-
Lin Feng Chen, Warner C. Greene, and Yajun Mu
- Subjects
Lysine ,Transcription Factor RelA ,Transcription factor complex ,Cell Cycle Proteins ,P300-CBP Transcription Factors ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Acetyltransferases ,p300-CBP Transcription Factors ,Molecular Biology ,Transcription factor ,Histone Acetyltransferases ,Cell Nucleus ,RELA ,General Immunology and Microbiology ,General Neuroscience ,NF-kappa B ,Acetylation ,Articles ,Molecular biology ,Cell biology ,IκBα ,Enhancer Elements, Genetic ,Mutation ,Transcription Factors - Abstract
The nuclear function of the heterodimeric NF-kappaB transcription factor is regulated in part through reversible acetylation of its RelA subunit. We now demonstrate that the p300 and CBP acetyltransferases play a major role in the in vivo acetylation of RelA, principally targeting lysines 218, 221 and 310 for modification. Analysis of the functional properties of hypoacetylated RelA mutants containing lysine-to-arginine substitutions at these sites and of wild-type RelA co-expressed in the presence of a dominantly interfering mutant of p300 reveals that acetylation at lysine 221 in RelA enhances DNA binding and impairs assembly with IkappaBalpha. Conversely, acetylation of lysine 310 is required for full transcriptional activity of RelA in the absence of effects on DNA binding and IkappaBalpha assembly. Together, these findings highlight how site-specific acetylation of RelA differentially regulates distinct biological activities of the NF-kappaB transcription factor complex.
- Published
- 2002
35. The NF-kappa B-inducing kinase induces PC12 cell differentiation and prevents apoptosis
- Author
-
Warner C. Greene, Erik D. Foehr, Lin Feng Chen, Romas Geleziunas, Alison O'Mahony, Noronha Carlos Manuel De, Xin Lin, and Jan Bohuslav
- Subjects
MAPK/ERK pathway ,Neurite ,Base Sequence ,Kinase ,Cellular differentiation ,Oligonucleotides ,Apoptosis ,Cell Differentiation ,Cell Biology ,IκB kinase ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,PC12 Cells ,Cell biology ,Rats ,IκBα ,Phosphorylation ,Animals ,Signal transduction ,Molecular Biology ,Signal Transduction - Abstract
NF-kappa B has been implicated in the survival and differentiation of PC12 cells. In this study, we examined the effect of the NF-kappa B-inducing kinase (NIK) on these processes. When inducibly expressed in PC12 cells, a kinase-proficient but not -deficient form of NIK promoted neurite process formation and mediated anti-apoptotic signaling. As expected, NIK expression led to I kappa B kinase activation and induced nuclear translocation of NF-kappa B. However, NIK-induced neurite outgrowth was only partially blocked by concomitant expression of a nondegradable form of I kappa B alpha that completely blocks NF-kappa B induction. In search of additional signaling pathways activated by NIK, we now demonstrate that NIK activates MEK1 phosphorylation and induces the Erk1/Erk2 MAPK pathway. Treatment of PC12 cells with PD98059, a MEK1 inhibitor, potently blocked neurite process formation; however, a dominantly interfering mutant of the upstream Shc adapter failed to alter this response. These findings reveal a new function for NIK as a MEK1-dependent activator of the MAPK pathway and implicate both the I kappa B kinase and MAPK signaling cascades in NIK-induced differentiation of PC12 cells.
- Published
- 2000
36. Intrinsic transcriptional activation-inhibition domains of the polyomavirus enhancer binding protein 2/core binding factor at subunit revealed in the presence of the β subunit
- Author
-
Tomohiko Kanno, Yoshiaki Ito, Lin Feng Chen, Woo-Young Kim, Yuka Kanno, and Eiko Ogawa
- Subjects
Gene Expression Regulation, Viral ,Transcriptional Activation ,Core Binding Factor alpha Subunits ,Protein subunit ,DNA Mutational Analysis ,Molecular Sequence Data ,Core Binding Factor Alpha 1 Subunit ,Biology ,Core binding factor ,Interleukin 10 receptor, alpha subunit ,Transactivation ,Jurkat Cells ,Mice ,Enhancer binding ,Animals ,Humans ,Nuclear Matrix ,Amino Acid Sequence ,Molecular Biology ,Transcription factor ,G alpha subunit ,Sequence Deletion ,Transcriptional Regulation ,Macrophage Colony-Stimulating Factor ,Cell Biology ,Molecular biology ,Cell Compartmentation ,Rats ,DNA-Binding Proteins ,Enhancer Elements, Genetic ,Transcription Factor AP-2 ,Polyomavirus ,Protein Binding ,Transcription Factors - Abstract
A member of the polyomavirus enhancer binding protein 2/core binding factor (PEBP2/CBF) is composed of PEBP2 alphaB1/AML1 (as the alpha subunit) and a beta subunit. It plays an essential role in definitive hematopoiesis and is frequently involved in the chromosomal abnormalities associated with leukemia. In the present study, we report functionally separable modular structures in PEBP2 alphaB1 for DNA binding and for transcriptional activation. DNA binding through the Runt domain of PEBP2 alphaB1 was hindered by the adjacent carboxy-terminal region, and this inhibition was relieved by interaction with the beta subunit. Utilizing a reporter assay system in which both the alpha and beta subunits are required to achieve strong transactivation, we uncovered the presence of transcriptional activation and inhibitory domains in PEBP2 alphaB1 that were only apparent in the presence of the beta subunit. The inhibitory domain keeps the full transactivation potential of full-length PEBP2 alphaB1 below its maximum potential. Fusion of the transactivation domain of PEBP2 alphaB1 to the yeast GAL4 DNA-binding domain conferred transactivation potential, but further addition of the inhibitory domain diminished the activity. These results suggest that the activity of the alpha subunit as a transcriptional activator is regulated intramolecularly as well as by the beta subunit. PEBP2 alphaB1 and the beta subunit were targeted to the nuclear matrix via signals distinct from the nuclear localization signal. Moreover, the transactivation domain by itself was capable of associating with the nuclear matrix, which implies the existence of a relationship between transactivation and nuclear matrix attachment.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.