Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Nat Immunol ; 12(1): 96-104, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151104

ABSTRACT

Overactive responses by interleukin 17 (IL-17)-producing helper T cells (T(H)17 cells) are tightly linked to the development of autoimmunity, yet the factors that negatively regulate the differentiation of this lineage remain unknown. Here we report that the transcription factor T-bet suppressed development of the T(H)17 cell lineage by inhibiting transcription of Rorc (which encodes the transcription factor RORγt). T-bet interacted with the transcription factor Runx1, and this interaction blocked Runx1-mediated transactivation of Rorc. T-bet Tyr304 was required for formation of the T-bet-Runx1 complex, for blockade of Runx1 activity and for inhibition of the T(H)17 differentiation program. Our data reinforce the idea of master regulators that shape immune responses by simultaneously activating one genetic program while silencing the activity of competing regulators in a common progenitor cell.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Box Domain Proteins/metabolism , Th17 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/immunology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation , Gene Regulatory Networks , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Protein Binding/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th17 Cells/immunology , Th17 Cells/pathology
2.
Cell Mol Life Sci ; 79(3): 186, 2022 Mar 13.
Article in English | MEDLINE | ID: mdl-35279781

ABSTRACT

Transcriptional co-activator with PDZ-binding motif (TAZ) is a key mediator of the Hippo signaling pathway and regulates structural and functional homeostasis in various tissues. TAZ activation is associated with the development of pancreatic cancer in humans, but it is unclear whether TAZ directly affects the structure and function of the pancreas. So we sought to identify the TAZ function in the normal pancreas. TAZ defect caused structural changes in the pancreas, particularly islet cell shrinkage and decreased insulin production and ß-cell markers expression, leading to hyperglycemia. Interestingly, TAZ physically interacted with the pancreatic and duodenal homeobox 1 (PDX1), a key insulin transcription factor, through the N-terminal domain of TAZ and the homeodomain of PDX1. TAZ deficiency decreased the DNA-binding and transcriptional activity of PDX1, whereas TAZ overexpression promoted PDX1 activity and increased insulin production even in a low glucose environment. Indeed, high glucose increased insulin production by turning off the Hippo pathway and inducing TAZ activation in pancreatic ß-cells. Ectopic TAZ overexpression along with PDX1 activation was sufficient to produce insulin in non-ß-cells. TAZ deficiency impaired the mesenchymal stem cell differentiation into insulin-producing cells (IPCs), whereas TAZ recovery restored normal IPCs differentiation. Compared to WT control, body weight increased in TAZ-deficient mice with age and even more with a high-fat diet (HFD). TAZ deficiency significantly exacerbated HFD-induced glucose intolerance and insulin resistance. Therefore, TAZ deficiency impaired pancreatic insulin production, causing hyperglycemia and exacerbating HFD-induced insulin resistance, indicating that TAZ may have a beneficial effect in treating insulin deficiency in diabetes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation , Cell Line , Diet, High-Fat , Glucose/pharmacology , Hippo Signaling Pathway/drug effects , Homeodomain Proteins/genetics , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperglycemia/veterinary , Insulin/genetics , Insulin Resistance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Trans-Activators/genetics , Transcriptional Activation
3.
J Cell Physiol ; 237(12): 4504-4516, 2022 12.
Article in English | MEDLINE | ID: mdl-36250997

ABSTRACT

Chronic liver injury follows inflammation and liver fibrosis; however, the molecular mechanism underlying fibrosis has not been fully elucidated. In this study, the role of ductal WW domain-containing transcription regulator 1 (WWTR1)/transcriptional coactivator with PDZ-binding motif (TAZ) was investigated after liver injury. Ductal TAZ-knockout (DKO) mice showed decreased liver fibrosis following a Diethyl 1,4-dihydro-2,4,6-trimethyl-3,5-pyridinedicarboxylate (DDC) diet compared to wild-type (WT) mice, as evidenced by decreased expression levels of fibrosis inducers, including connective tissue growth factor (Ctgf)/cellular communication network factor 2 (CCN2), cysteine-rich angiogenic inducer 61 (Cyr61/CCN1), and transforming growth factor beta 1 (Tgfb1), in DKO mice. Similarly, TAZ-knockout (KO) cholangiocyte organoids showed decreased expression of fibrosis inducers. Additionally, the culture supernatant of TAZ-KO cholangiocyte organoids decreased the fibrogenic gene expression in liver stellate cells. Further studies revealed that prominin 1 (PROM1/CD133) stimulated TAZ for fibrosis. After the administration of DDC diet, fibrosis was decreased in CD133-KO (CD133-KO) mice compared to that in WT mice. Similarly, CD133-KO cholangiocyte organoids showed decreased Ctgf, Cyr61, and Tgfb1 expression levels compared to WT cholangiocyte organoids. Mechanistically, CD133 stabilized TAZ via Src activation. Inhibition of Src decreased TAZ levels. Similarly, CD133-knockdown HCT116 cells showed decreased TAZ levels, but reintroduction of active Src recovered the TAZ levels. Taken together, our results suggest that TAZ facilitates liver fibrosis after a DDC diet via the CD133-Src-TAZ axis.


Subject(s)
Adaptor Proteins, Signal Transducing , Chemical and Drug Induced Liver Injury, Chronic , Trans-Activators , Animals , Mice , Diet , Fibrosis , Intracellular Signaling Peptides and Proteins , Liver , Liver Cirrhosis/chemically induced , Mice, Knockout , Transcription Factors/genetics , Proto-Oncogene Proteins pp60(c-src) , Adaptor Proteins, Signal Transducing/genetics
4.
Proc Natl Acad Sci U S A ; 116(8): 3106-3111, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30718407

ABSTRACT

Expression of ectonucleotidase CD39 contributes to the suppressive activity of Foxp3+ regulatory T cells (Tregs) by hydrolyzing immunogenic ATP into AMP. The molecular mechanism that drives CD39 expression on Tregs remains elusive. We found that tumor-infiltrating Tregs (Ti-Tregs) failed to up-regulate CD39 in mice lacking EBI3 subunit of IL-27 or IL-27Ra. Mixed bone marrow chimera and in vitro studies showed that IL-27 signaling in Tregs directly drives CD39 expression on Ti-Tregs in a STAT1-dependent, but STAT3- and T-bet-independent, manner. Tregs stimulated with IL-27 showed enhanced suppressive activities against CD8+ T cell responses in vitro. Moreover, IL-27Ra-deficient Tregs and STAT1-deficient Tregs were less efficient than WT Tregs in suppressing antitumor immunity in vivo. CD39 inhibition significantly abolished IL-27-induced suppressive activities of Tregs. Thus, IL-27 signaling in Tregs critically contributes to protumorigenic properties of Tregs via up-regulation of CD39.


Subject(s)
Antigens, CD/genetics , Apyrase/genetics , Carcinogenesis/genetics , Forkhead Transcription Factors/genetics , STAT1 Transcription Factor/genetics , Animals , Gene Expression Regulation, Neoplastic , Humans , Interleukin-27/genetics , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th17 Cells
5.
J Lipid Res ; 62: 100152, 2021.
Article in English | MEDLINE | ID: mdl-34808194

ABSTRACT

Testosterone is a hormone essential for male reproductive function. It is produced primarily by Leydig cells in the testicle through activation of steroidogenic acute regulatory protein and a series of steroidogenic enzymes, including a cytochrome P450 side-chain cleavage enzyme (cytochome P450 family 11 subfamily A member 1), 17α-hydroxylase (cytochrome P450 family 17 subfamily A member 1), and 3ß-hydroxysteroid dehydrogenase. These steroidogenic enzymes are mainly regulated at the transcriptional level, and their expression is increased by the nuclear receptor 4A1. However, the effect on Leydig cell function of a small molecule-activating ligand, amodiaquine (AQ), is unknown. We found that AQ effectively and significantly increased testosterone production in TM3 and primary Leydig cells through enhanced expression of steroidogenic acute regulatory protein, cytochome P450 family 11 subfamily A member 1, cytochrome P450 family 17 subfamily A member 1, and 3ß-hydroxysteroid dehydrogenase. Concurrently, AQ dose-dependently increased the expression of 3-hydroxy-3-methylglutaryl-CoA reductase, a key enzyme in the cholesterol synthesis pathway, through induction of the transcriptional and DNA-binding activities of nuclear receptor 4A1, contributing to increased cholesterol synthesis in Leydig cells. Furthermore, AQ increased the expression of fatty acid synthase and diacylglycerol acyltransferase and potentiated de novo synthesis of fatty acids and triglycerides (TGs). Lipidomics profiling further confirmed a significant elevation of intracellular lipid and TG levels by AQ in Leydig cells. These results demonstrated that AQ effectively promotes testosterone production and de novo synthesis of cholesterol and TG in Leydig cells, indicating that AQ may be beneficial for treating patients with Leydig cell dysfunction and subsequent testosterone deficiency.


Subject(s)
Amodiaquine/pharmacology , Cholesterol/biosynthesis , Leydig Cells/drug effects , Testosterone/biosynthesis , Triglycerides/biosynthesis , Animals , Leydig Cells/metabolism , Male , Mice , Mice, Inbred C57BL
6.
FASEB J ; 34(4): 5332-5347, 2020 04.
Article in English | MEDLINE | ID: mdl-32067268

ABSTRACT

Transcriptional coactivator with PDZ-binding motif (TAZ) plays crucial role in maintaining testicular structure and function via regulation of senescence of spermatogenic cells. However, it remains unclear whether TAZ is involved in testosterone biosynthesis in testicular Leydig cells. We found that TAZ deficiency caused aberrant Leydig cell expansion and increased lipid droplet formation, which was significantly associated with increased lipogenic enzyme expression. Additionally, the expression of key steroidogenic enzymes, including steroidogenic acute regulatory protein, cytochrome P450 (CYP) 11A1, CYP17A1, and 3ß-hydroxysteroid dehydrogenase, was greatly increased in TAZ-deficient testes and primary Leydig cells. Interestingly, the transcriptional activity of nuclear receptor 4 A1 (NR4A1) was dramatically suppressed by TAZ; however, the protein expression and the subcellular localization of NR4A1 were not affected by TAZ. TAZ directly associated with the N-terminal region of NR4A1 and substantially suppressed its DNA-binding and transcriptional activities. Stable expression of TAZ in the mouse Leydig TM3 cell line decreased the expression of key steroidogenic enzymes, whereas knockdown of endogenous TAZ in TM3 cells increased transcripts of steroidogenic genes induced by NR4A1. Consistently, testosterone production was enhanced within TAZ-deficient Leydig cells. However, TAZ deficiency resulted in decreased testosterone secretion caused by dysfunctional mitochondria and lysosomes. Therefore, TAZ plays essential role in NR4A1-induced steroidogenic enzyme expression and testosterone production in Leydig cells.


Subject(s)
17-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Leydig Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Phosphoproteins/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Testosterone/metabolism , Trans-Activators/physiology , 17-Hydroxysteroid Dehydrogenases/genetics , 17-Hydroxysteroid Dehydrogenases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism
7.
Biochem Biophys Res Commun ; 524(1): 242-248, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31983436

ABSTRACT

Ultraviolet (UV) irradiation induces the proliferation and differentiation of keratinocytes in the basal layer of the epidermis, which increases epidermal thickness in skin regeneration. However, the mechanism underlying this phenomenon is not yet known in detail. In this study, we aimed to demonstrate that the transcriptional coactivator with PDZ-binding motif (TAZ) stimulates epidermal regeneration by increasing keratinocyte proliferation. During epidermal regeneration, TAZ is localized in the nucleus of keratinocytes of the basal layer and stimulates epidermal growth factor receptor (EGFR) signaling. TAZ depletion in keratinocytes decreased EGFR signaling activation, which delays epidermal regeneration. Interestingly, TAZ stimulated the transcription of amphiregulin (AREG), a ligand of EGFR, through TEAD-mediated transcriptional activation. Together, these results show that TAZ stimulates EGFR signaling through AREG induction, suggesting that it plays an important role in epidermal regeneration.


Subject(s)
Amphiregulin/genetics , Epidermis/physiology , Regeneration , Trans-Activators/metabolism , Transcription, Genetic , Ultraviolet Rays , Adaptor Proteins, Signal Transducing , Amphiregulin/metabolism , Animals , Cell Proliferation/radiation effects , Epidermis/radiation effects , ErbB Receptors/metabolism , Gene Deletion , Humans , Keratinocytes/metabolism , Keratinocytes/radiation effects , Ligands , Male , Mice, Inbred C57BL , Mice, Knockout , Regeneration/radiation effects , Signal Transduction/radiation effects , Transcription, Genetic/radiation effects
8.
FASEB J ; 33(10): 11381-11395, 2019 10.
Article in English | MEDLINE | ID: mdl-31311305

ABSTRACT

We previously reported that syndecan-2 expression is increased on the colonic epithelium during chronic inflammation. Here, we report that syndecan-2 exhibits a different pattern of site-specific colonic expression during acute inflammation. Syndecan-2 expression was up-regulated predominantly in the proximal colon of dextran sulfate sodium-induced colitis mice. The colitis-associated up-regulation of syndecan-2 was barely detected in Rag-1-/- (recombination activating gene 1 knockout) mice under colitis-inducing conditions. Increased syndecan-2 expression correlated with increased levels of infiltrated CD4+ IL-17A+ T cells in the proximal colon. Serum levels of IL-17A were increased during the acute inflammatory response in normal mice but not Rag-1-/- mice. IL-17A directly induced IL-17 receptor (IL-17RA) and syndecan-2 expression in ex vivo-cultured proximal colon tissues and adenoma cell lines from proximal colon. IL-17RA knockdown reduced the IL-17A-mediated syndecan-2 expression in SNU1235 cells. No elevation of syndecan-2 or IL-17RA was observed in colonic tissues from IL-17A-/- mice during colitis induction. Finally, increased expression of syndecan-2 and IL-17RA was observed in the proximal colons of cecal ligation and puncture-induced sepsis mice and infectious pan colitis patients. Together, these data suggest that acute inflammation induces syndecan-2 expression predominantly in the proximal colon via IL-17A-IL-17RA signaling during the early stage of the inflammatory response and that proximal colonic syndecan-2 might be a biomarker for acute inflammation.-Hong, H., Song, H.-K., Hwang, E. S., Lee, A. R., Han, D. S., Kim, S.-E., Oh, E.-S. Up-regulation of syndecan-2 in proximal colon correlates with acute inflammation.


Subject(s)
Colon/metabolism , Inflammation/metabolism , Syndecan-2/metabolism , Up-Regulation/physiology , Animals , Cell Line, Tumor , Colitis/chemically induced , Colitis/metabolism , Colon/drug effects , Dextran Sulfate/pharmacology , Humans , Inflammation/chemically induced , Interleukin-17/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin-17/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology
9.
FASEB J ; 33(5): 5914-5923, 2019 05.
Article in English | MEDLINE | ID: mdl-30742777

ABSTRACT

In response to liver injury, the liver undergoes a regeneration process to retain its mass and function. However, the regeneration mechanism has not been fully clarified. This study investigated the role of transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo-signaling effector, in liver regeneration. We observed that TAZ stimulates liver regeneration after liver injury. After partial hepatectomy (PHx) or carbon tetrachloride damage, TAZ was required for liver regeneration to increase hepatic cell proliferation and resist hepatic apoptosis, which were decreased in liver-specific TAZ knockout (LKO) mice. TAZ stimulated macrophage infiltration, resulting in IL-6 production, which induced liver regeneration. In LKO mice, IL-6-induced activation of signal transducer and activator of transcription 3, ERK, and PKB was decreased. We also observed that periductal fibrogenesis was significantly increased in LKO mice during liver regeneration after PHx, which was caused by increased hepatic apoptosis. Our results suggest that TAZ stimulates liver regeneration through IL-6-induced hepatocyte proliferation and inhibition of cell death after liver injury.-Kim, A. R., Park, J. I., Oh, H. T., Kim, K. M., Hwang, J.-H., Jeong, M. G., Kim, E.-H., Hwang, E. S., Hong, J.-H. TAZ stimulates liver regeneration through interleukin-6-induced hepatocyte proliferation and inhibition of cell death after liver injury.


Subject(s)
Interleukin-6/metabolism , Liver Regeneration , Liver/injuries , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing , Alleles , Animals , Apoptosis , Carbon Tetrachloride , Cell Death , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatectomy , Hepatocytes/cytology , Hepatocytes/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism
10.
Proc Natl Acad Sci U S A ; 113(43): E6572-E6581, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27791029

ABSTRACT

The transcription factor T-bet (Tbox protein expressed in T cells) is one of the master regulators of both the innate and adaptive immune responses. It plays a central role in T-cell lineage commitment, where it controls the TH1 response, and in gene regulation in plasma B-cells and dendritic cells. T-bet is a member of the Tbox family of transcription factors; however, T-bet coordinately regulates the expression of many more genes than other Tbox proteins. A central unresolved question is how T-bet is able to simultaneously recognize distant Tbox binding sites, which may be located thousands of base pairs away. We have determined the crystal structure of the Tbox DNA binding domain (DBD) of T-bet in complex with a palindromic DNA. The structure shows a quaternary structure in which the T-bet dimer has its DNA binding regions splayed far apart, making it impossible for a single dimer to bind both sites of the DNA palindrome. In contrast to most other Tbox proteins, a single T-bet DBD dimer binds simultaneously to identical half-sites on two independent DNA. A fluorescence-based assay confirms that T-bet dimers are able to bring two independent DNA molecules into close juxtaposition. Furthermore, chromosome conformation capture assays confirm that T-bet functions in the direct formation of chromatin loops in vitro and in vivo. The data are consistent with a looping/synapsing model for transcriptional regulation by T-bet in which a single dimer of the transcription factor can recognize and coalesce distinct genetic elements, either a promoter plus a distant regulatory element, or promoters on two different genes.


Subject(s)
Chromatin/chemistry , DNA/chemistry , Genome , T-Box Domain Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , Chromatin/metabolism , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Inverted Repeat Sequences , Mice , Models, Molecular , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Xenopus laevis
11.
Biochem Biophys Res Commun ; 487(2): 409-414, 2017 05 27.
Article in English | MEDLINE | ID: mdl-28419840

ABSTRACT

Bone mass is controlled by a balance between bone resorption and formation by osteoclasts and osteoblasts, respectively. An imbalance between osteoblasts and osteoclasts increases the risk of osteoporosis and fractures. Although inhibition of osteoclasts is beneficial for preventing and treating osteoporosis, enhanced bone formation through activation of osteoblast differentiation can be a more promising therapeutic approach. In this study, we attempted to isolate small molecules that promote osteoblast differentiation and found that IBIP (3-(2,3-dimethoxyphenyl)-1-[9-methyl-2-phenyl-9H-imidazo[1,2-a]benzimidazol-3-yl]-2-propen-1-one) was a potent activator of osteoblast differentiation. Upon bone morphogenetic protein-2 (BMP2) stimulation, IBIP promoted osteoblast differentiation and increased the expression of osteoblast-specific gene markers, such as osterix and alkaline phosphatase, in a dose-dependent manner. The phosphorylation of SMADs and extracellular signal-regulated kinase (ERK) increased after IBIP treatment. While enhanced SMAD phosphorylation by IBIP was abolished by a BMP inhibitor, IBIP-induced ERK phosphorylation was sustained in the presence of this inhibitor, but was decreased by an ERK kinase inhibitor. Suppression of IBIP-induced SMAD and ERK phosphorylation diminished osteoblast differentiation. Most importantly, IBIP enhanced bone formation and calcification in a BMP2-independent manner in vitro and advanced the skeletal development of zebrafish larvae in vivo. Collectively, IBIP may have beneficial effects on bone loss through potentiation of bone formation.


Subject(s)
Benzimidazoles/administration & dosage , Bone Development/physiology , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/physiology , 3T3 Cells , Animals , Bone Development/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Imidazoles/administration & dosage , Mice , Osteoblasts/cytology , Osteogenesis/drug effects , Zebrafish
12.
Biochem Biophys Res Commun ; 486(2): 378-384, 2017 04 29.
Article in English | MEDLINE | ID: mdl-28315325

ABSTRACT

Muscle loss is a typical process of aging. Green tea consumption is known to slow down the progress of aging. Their underlying mechanisms, however, remain largely unknown. In this study, we investigated the effect of (-)-epigallocatechin-3-gallate (EGCG), a polyphenolic compound of green tea, on myogenic differentiation and found that EGCG significantly increases myogenic differentiation. After EGCG treatment, the expression of myogenic marker genes, such as myosin heavy chain, are increased through activation of TAZ, a transcriptional coactivator with a PDZ-binding motif. TAZ-knockdown does not stimulate EGCG-induced myogenic differentiation. EGCG facilitates the interaction between TAZ and MyoD, which stimulates MyoD-mediated gene transcription. EGCG induces nuclear localization of TAZ through the dephosphorylation of TAZ at its Ser89 residue, which relieves 14-3-3 binding in the cytosol. Interestingly, inactivation of Lats kinase is observed after EGCG treatment, which is responsible for the production of dephosphorylated TAZ. Together, these results suggest that EGCG induces myogenic differentiation through TAZ, suggesting that TAZ plays an important role in EGCG induced muscle regeneration.


Subject(s)
Catechin/analogs & derivatives , Cell Differentiation/drug effects , Myoblasts/drug effects , Satellite Cells, Skeletal Muscle/drug effects , Transcription Factors/agonists , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Acyltransferases , Animals , Catechin/pharmacology , Cell Line , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Primary Cell Culture , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction , Tea/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
13.
Biochem Biophys Res Commun ; 489(2): 142-148, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28546002

ABSTRACT

Muscle weakness is one of the most common symptoms in aged individuals and increases risk of mortality. Thus, maintenance of muscle mass is important for inhibiting aging. In this study, we investigated the effect of catechins, polyphenol compounds in green tea, on muscle regeneration. We found that (-)-epicatechin gallate (ECG) and (-)-epigallocatechin-3-gallate (EGCG) activate satellite cells by induction of Myf5 transcription factors. For satellite cell activation, Akt kinase was significantly induced after ECG treatment and ECG-induced satellite cell activation was blocked in the presence of Akt inhibitor. ECG also promotes myogenic differentiation through the induction of myogenic markers, including Myogenin and Muscle creatine kinase (MCK), in satellite and C2C12 myoblast cells. Finally, EGCG administration to mice significantly increased muscle fiber size for regeneration. Taken together, the results suggest that catechins stimulate muscle stem cell activation and differentiation for muscle regeneration.


Subject(s)
Catechin/pharmacology , Muscles/drug effects , Muscles/physiology , Myogenic Regulatory Factor 5/biosynthesis , Regeneration/drug effects , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Animals , Catechin/chemistry , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Molecular Conformation , Muscles/cytology , Myogenic Regulatory Factor 5/metabolism , Structure-Activity Relationship
14.
Bioorg Med Chem ; 25(12): 3127-3134, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28442260

ABSTRACT

A growing number of studies have demonstrated that interleukin (IL)-6 plays pathological roles in the development of chronic inflammatory disease and autoimmune disease by activating innate immune cells and by stimulating adaptive inflammatory T cells. So, suppression of IL-6 function may be beneficial for prevention and treatment of chronic inflammatory disease. This study reports that a series of synthetic derivatives of benzoxazole have suppressive effects on IL-6-mediated signaling. Among 16 synthetic derivatives of benzoxazole, the compounds 4, 6, 11, 15, 17, and 19 showed a strong suppressive activity against IL-6-induced phosphorylation of signal transducer and activator of transcription (STAT) 3 by 80-90%. While the cell viability was strongly decreased by compounds 11, 17, 19, the compounds 4, 6, and 15 revealed less cytotoxicity. We then examined the effects of the compounds on inflammatory cytokine production by CD4+ T cells. CD4+ T cells were induced to differentiate into interferon (IFN)-γ-, IL-17-, or IL-4-producing effector T cells in the presence of either the compound 4 or the compound 7. While the inactive compound 7 had no significant effect on the cytokine production by effector T cells, the active compound 4 strongly suppressed the production of inflammatory cytokines IFN-γ and IL-17, and also inhibited allergic inflammatory cytokines IL-4, IL-5, and IL-13 produced by effector Th2 cells. These results suggest that a benzoxazole derivative, compound 4 effectively suppresses IL-6-STAT3 signaling and inflammatory cytokine production by T cells and provides a beneficial effect for treating chronic inflammatory and autoimmune disease.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Interleukin-6/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemical synthesis , Benzoxazoles/chemical synthesis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Hep G2 Cells , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-4/immunology , Interleukin-5/immunology , Interleukin-6/immunology , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , Signal Transduction/drug effects
15.
Biochem Biophys Res Commun ; 474(2): 345-350, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27109480

ABSTRACT

Chloroquine (CQ) and amodiaquine (AQ) have been used for treating or preventing malaria for decades, and their application has expanded into treating inflammatory disease in humans. CQ and AQ are applicable for controlling rheumatoid arthritis, but their molecular mechanisms of anti-inflammatory activity remain to be elucidated. In this study, we examined the effects of CQ and AQ on T cell activation and T cell-mediated immune response. CQ had no significant effect on T cell numbers, but decreased the population of T cells with a high division rate. However, AQ treatment significantly increased the number of cells with low division rates and eliminated cells with high division rates, resulting in the inhibition of T cell proliferation triggered by T cell receptor stimulation, of which inhibition occurred in developing effector T helper and regulatory T cells, regardless of the different exogenous cytokines. Interestingly, the cyclin-dependent kinase inhibitor p21 was significantly and dose-dependently increased by CQ, and more potently by AQ, while other cell cycle regulators were unchanged. Both CQ and AQ elevated the transcription level of p21 though the activation of p53, but also blocked p21 protein degradation in the presence of cycloheximide, causing p21 protein accumulation mainly in the nucleus. Sustained treatment of developing T cells with either CQ or AQ suppressed IFN-γ production in a dose dependent manner and potently inhibited the differentiation of IFN-γ-producing Th1 cells. These results demonstrate that CQ and AQ increase the expression level of p21 and inhibit T cell proliferation and the development of IFN-γ-producing Th1 cells, thereby revealing beneficial roles in treating a wide range of chronic inflammatory diseases mediated by inflammatory T cells.


Subject(s)
Amodiaquine/administration & dosage , Chloroquine/administration & dosage , Cyclin-Dependent Kinase Inhibitor p21/metabolism , T-Lymphocytes/physiology , Th1 Cells/cytology , Th1 Cells/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , Th1 Cells/drug effects , Treatment Outcome
16.
J Biol Chem ; 289(14): 9926-35, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24515112

ABSTRACT

Osteoporosis is a degenerative bone disease characterized by low bone mass and is caused by an imbalance between osteoblastic bone formation and osteoclastic bone resorption. It is known that the bioactive compounds present in green tea increase osteogenic activity and decrease the risk of fracture by improving bone mineral density. However, the detailed mechanism underlying these beneficial effects has yet to be elucidated. In this study, we investigated the osteogenic effect of (-)-epicatechin gallate (ECG), a major bioactive compound found in green tea. We found that ECG effectively stimulates osteoblast differentiation, indicated by the increased expression of osteoblastic marker genes. Up-regulation of osteoblast marker genes is mediated by increased expression and interaction of the transcriptional coactivator with PDZ-binding motif (TAZ) and Runt-related transcription factor 2 (RUNX2). ECG facilitates nuclear localization of TAZ through PP1A. PP1A is essential for osteoblast differentiation because inhibition of PP1A activity was shown to suppress ECG-mediated osteogenic differentiation. Taken together, the results showed that ECG stimulates osteoblast differentiation through the activation of TAZ and RUNX2, revealing a novel mechanism for green tea-stimulated osteoblast differentiation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Catechin/analogs & derivatives , Cell Differentiation/drug effects , Cell Nucleus/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Transcription Factors/biosynthesis , Transcriptional Activation/drug effects , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Acyltransferases , Animals , Catechin/pharmacology , Cell Differentiation/physiology , Cell Line , Cell Nucleus/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Humans , Mice , Osteoblasts/cytology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Transcription Factors/genetics , Transcriptional Activation/physiology
17.
Stem Cells ; 32(9): 2443-53, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24805773

ABSTRACT

We reported a functional incompetence in mesenchymal stem cells (MSCs) under uremia, but the mechanisms have not been explored. To study the mechanisms of dysfunctional MSCs induced by uremia, we characterized insulin signaling in MSCs and investigated the effect of uremic toxin, p-cresol, on the proangiogenic actions of insulin. In MSCs, insulin induced hypoxia-inducible factor (HIF)-1α, vascular endothelial growth factor, and stromal cell-derived factor 1α expressions via PI3K/Akt-dependent pathway. MSCs treated with p-cresol exhibited altered insulin signaling in a selective manner for insulin receptor substrate-1/PI3K/Akt pathway, whereas ERK pathway remained active. The insulin-induced increase of HIF-1α was blunted by p-cresol treatment. This Akt-selective insulin resistance was also observed in MSCs isolated from chronic kidney disease (CKD) mice. In mice model of hindlimb ischemia, blood flow recovery, capillary density, and local production of angiogenic factors in the ischemic limb treated with CKD MSCs were significantly inferior to those promoted by control MSCs. However, modifying CKD MSCs by overexpression of HIF-1α restored all of these changes. Taken together, these data suggest that p-cresol contributes to insulin resistance in a selective manner for Akt pathway. This might be a biological explanation for the functional incompetence of MSCs under uremia through defects in the insulin-induced elevation of HIF-1α protein expression.


Subject(s)
Bone Marrow/metabolism , Cresols/pharmacology , Insulin Resistance , Mesenchymal Stem Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Uremia/pathology , Animals , Cell Differentiation/drug effects , Cells, Cultured , HEK293 Cells , Humans , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Transfection , Uremia/metabolism
18.
J Immunol ; 190(11): 5764-70, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23616576

ABSTRACT

A T-box-containing protein expressed in T cells (T-bet) is a key transcription factor involved in the regulation of Th cell differentiation. Although T-bet-deficient CD4(+) T cells fail to produce IFN-γ and typically differentiate into Th2 cells in vitro, ectopic overexpression of T-bet elevates IFN-γ and suppresses production of IL-2 and Th2 cytokines through different mechanisms. Despite the importance of the T-bet protein level, the regulatory mechanisms that control T-bet protein stability are largely unknown. In this study, we found that T-bet underwent proteasomal degradation via ubiquitination at Lys-313. Despite its robust accumulation following lysine mutation, T-bet(K313R) failed to increase IFN-γ production because of diminished DNA binding activity, as demonstrated in the crystal structure of T-bet-DNA complex. Strikingly, T-bet(K313R) entirely lost the ability to suppress IL-2 production and Th2 cell development; this was due to loss of its interaction with NFAT1. We further identified that the T-bet(K313R) reduced the phosphorylation of T-bet at Thr-302, and that threonine phosphorylation was essential for T-bet interaction with NFAT1 and suppression of NFAT1 activity. Retroviral transduction of T-bet(T302A) into T-bet-deficient cells restored IFN-γ levels compared with those induced by wild-type T-bet, but this mutant failed to inhibit IL-2 and Th2 cytokine production. Collectively, these data show that Lys-313 in the T-box domain is essential for controlling T-bet protein stability via ubiquitin-dependent degradation, T-bet binding to the IFN-γ promoter, and for the interaction with and suppression of NFAT1. Thus, multiple posttranslational modifications of T-bet are involved in fine-tuning cytokine production during Th cell development.


Subject(s)
Lysine/metabolism , T-Box Domain Proteins/chemistry , T-Box Domain Proteins/metabolism , Threonine/metabolism , Amino Acid Sequence , Animals , Binding Sites , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Humans , Interferon-gamma/genetics , Mice , Mice, Knockout , Molecular Docking Simulation , Molecular Sequence Data , Mutation , NFATC Transcription Factors/metabolism , Phosphorylation , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Conformation , Protein Stability , Proteolysis , Sequence Alignment , T-Box Domain Proteins/genetics , Th2 Cells/metabolism , Transcription, Genetic , Ubiquitination
19.
J Immunol ; 191(8): 4029-37, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048895

ABSTRACT

Peroxiredoxin (Prx) II is an intracellular antioxidant molecule that eliminates hydrogen peroxide, employing a high substrate-binding affinity. PrxII deficiency increases the levels of intracellular reactive oxygen species in many types of cells, which may increase reactive oxygen species-mediated inflammation. In this study, we investigated the susceptibility of PrxII knockout (KO) mice to experimentally induced colitis and the effects of PrxII on the immune system. Wild-type mice displayed pronounced weight loss, high mortality, and colon shortening after dextran sulfate sodium administration, whereas colonic inflammation was significantly attenuated in PrxII KO mice. Although macrophages were hyperactivated in PrxII KO mice, the amount of IFN-γ and IL-17 produced by CD4(+) T cells was substantially reduced. Foxp3(+) regulatory T (Treg) cells were elevated, and Foxp3 protein expression was increased in the absence of PrxII in vitro and in vivo. Restoration of PrxII into KO cells suppressed the increased Foxp3 expression. Interestingly, endogenous PrxII was inactivated through hyperoxidation during Treg cell development. Furthermore, PrxII deficiency stabilized FoxO1 expression by reducing mouse double minute 2 homolog expression and subsequently activated FoxO1-mediated Foxp3 gene transcription. PrxII overexpression, in contrast, reduced FoxO1 and Foxp3 expression. More interestingly, adoptive transfer of naive CD4(+) T cells from PrxII KO mice into immune-deficient mice attenuated T cell-induced colitis, with a reduction in mouse double minute 2 homolog expression and an increase in FoxO1 and Foxp3 expression. These results suggest that inactivation of PrxII is important for the stability of FoxO1 protein, which subsequently mediates Foxp3(+) Treg cell development, thereby attenuating colonic inflammation.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , Peroxiredoxins/metabolism , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/genetics , Dextran Sulfate , Forkhead Box Protein O1 , Interferon-gamma/metabolism , Interleukin-17/metabolism , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Peroxiredoxins/genetics , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/metabolism
20.
J Immunol ; 188(5): 2244-53, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22266280

ABSTRACT

IL-10 is a multifunctional cytokine that plays a crucial role in immunity and tolerance. IL-10 is produced by diverse immune cell types, including B cells and subsets of T cells. Although Th1 produce IL-10, their expression levels are much lower than Th2 cells under conventional stimulation conditions. The potential role of E26 transformation-specific 1 (Ets-1) transcription factor as a negative regulator for Il10 gene expression in CD4(+) T cells has been implicated previously. In this study, we investigated the underlying mechanism of Ets-1-mediated Il10 gene repression in Th1 cells. Compared with wild type Th1 cells, Ets-1 knockout Th1 cells expressed a significantly higher level of IL-10, which is comparable with that of wild type Th2 cells. Upregulation of IL-10 expression in Ets-1 knockout Th1 cells was accompanied by enhanced chromatin accessibility and increased recruitment of histone H3 acetylation at the Il10 regulatory regions. Reciprocally, Ets-1 deficiency significantly decreased histone deacetylase 1 (HDAC1) enrichment at the Il10 regulatory regions. Treatment with trichostatin A, an inhibitor of HDAC family, significantly increased Il10 gene expression by increasing histone H3 acetylation recruitment. We further demonstrated a physical interaction between Ets-1 and HDAC1. Coexpression of Ets-1 with HDAC1 synergistically repressed IL-10 transcription activity. In summary, our data suggest that an interaction of Ets-1 with HDAC1 represses the Il10 gene expression in Th1 cells.


Subject(s)
Down-Regulation/immunology , Gene Expression Regulation/immunology , Histone Deacetylase 1/physiology , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Proto-Oncogene Protein c-ets-1/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Down-Regulation/genetics , HEK293 Cells , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Humans , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Protein c-ets-1/deficiency , Proto-Oncogene Protein c-ets-1/metabolism , Th1 Cells/cytology , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL