Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Genes Cells ; 25(4): 232-241, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31991027

ABSTRACT

Previously, we found that the basic helix-loop-helix transcriptional repressor DEC1 interacts with the PPARγ:RXRα heterodimer, a master transcription factor for adipogenesis and lipogenesis, to suppress transcription from PPARγ target genes (Noshiro et al., Genes to Cells, 2018, 23:658-669). Because the expression of PPARγ and several of its target genes exhibits circadian rhythmicity in white adipose tissue (WAT), we examined the expression profiles of PPARγ target genes in wild-type and Dec1-/- mice. We found that the expression of PPARγ target genes responsible for lipid metabolism, including the synthesis of triacylglycerol from free fatty acids (FFAs), lipid storage and the lipolysis of triacylglycerol to FFAs, oscillates in a circadian manner in WAT. Moreover, DEC1 deficiency led to a marked increase in the expression of these genes at night (Zeitgeber times 16 and 22), resulting in disruption of circadian rhythms. Serum FFA levels in wild-type mice also showed circadian oscillations, but these were disrupted by DEC1 deficiency, leading to reduced FFA levels. These results suggest that PPARγ:RXRα and DEC1 cooperatively generate the circadian expression of PPARγ target genes through PPAR-responsive elements in WAT.


Subject(s)
Adipose Tissue, White/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Circadian Rhythm/genetics , Homeodomain Proteins/metabolism , Lipid Metabolism , PPAR gamma/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Fatty Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/metabolism
2.
Hypertension ; 72(3): 746-754, 2018 09.
Article in English | MEDLINE | ID: mdl-30012868

ABSTRACT

Blood pressure shows a circadian rhythm, and recent studies have suggested the involvement of a molecular clock system in its control. In the clock system, the CLOCK (circadian locomotor output cycles kaput):BMAL1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like protein-1) heterodimer enhances promoter activity of clock genes, and DEC1 (BHLHE40/STRA13/SHARP-2) represses CLOCK/BMAL1-enhanced promoter activity through competition for binding to the clock element, CACGTG E-box. However, the molecular mechanisms by which this system regulates blood pressure remain unclear. Here, we show that DEC1 suppressed the expression of ATP1B1, which encodes the ß1 subunit of the Na+/K+-ATPase and elevated blood pressure. Using chromatin immunoprecipitation and chromatin immunoprecipitation-on-chip analyses, we found that DEC1 and CLOCK bound to E-boxes in the ATP1B1 promoter. Luciferase assays revealed that CLOCK:BMAL1 heterodimer enhanced transcription from the ATP1B1 promoter, whereas DEC1 suppressed this transactivation. Accordingly, Atp1b1 mRNA and protein levels in mouse kidney, aorta, and heart showed a circadian rhythm that was antiphasic to the blood pressure rhythm. Furthermore, Dec1-deficient mice showed enhanced Atp1b1 expression in these tissues and reduced blood pressure. In contrast, Clock-mutant mice showed reduced Atp1b1 expression and elevated blood pressure. Our results raise the possibility that transcriptional regulation of Atp1b1 by DEC1 and CLOCK:BMAL1 contributes to blood pressure.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Blood Pressure/genetics , CLOCK Proteins/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Sodium-Potassium-Exchanging ATPase/genetics , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Pressure/physiology , CLOCK Proteins/metabolism , Cells, Cultured , Circadian Rhythm , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , Sodium-Potassium-Exchanging ATPase/metabolism
3.
Genes Cells ; 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29968353

ABSTRACT

Obesity is a major public health problem in developed countries resulting from increased food intake and decreased energy consumption and usually associated with abnormal lipid metabolism. Here, we show that DEC1, a basic helix-loop-helix transcription factor, plays an important role in the regulation of lipid consumption in mouse brown adipose tissue (BAT), which is the major site of thermogenesis. Homozygous Dec1 deletion attenuated high-fat-diet-induced obesity, adipocyte hypertrophy, fat volume and hepatic steatosis. Furthermore, DEC1 deficiency increased body temperature during daytime and enhanced the expression of uncoupler protein 1, a key factor of thermogenesis, and various lipolysis-related genes in interscapular BAT. In vitro experiments suggested that DEC1 suppresses the expression of various lipolysis-related genes induced by the heterodimer of peroxisome proliferator-activated receptor γ and retinoid X receptor α (RXRα) through direct binding to RXRα. These observations suggest that enhanced lipolysis in BAT caused by DEC1 deficiency leads to an increase in lipid consumption, thereby decreasing lipid accumulation in adipose tissues and the liver. Thus, DEC1 may serve as an energy-saving factor that suppresses lipid consumption, which may be relevant to managing obesity.

4.
Stem Cells Int ; 2016: 8035759, 2016.
Article in English | MEDLINE | ID: mdl-27648077

ABSTRACT

Msh homeobox 1 (MSX1) encodes a transcription factor implicated in embryonic development of limbs and craniofacial tissues including bone and teeth. Although MSX1 regulates osteoblast differentiation in the cranial bone of young animal, little is known about the contribution of MSX1 to the osteogenic potential of human cells. In the present study, we investigate the role of MSX1 in osteogenic differentiation of human dental pulp stem cells isolated from deciduous teeth. When these cells were exposed to osteogenesis-induction medium, runt-related transcription factor-2 (RUNX2), bone morphogenetic protein-2 (BMP2), alkaline phosphatase (ALPL), and osteocalcin (OCN) mRNA levels, as well as alkaline phosphatase activity, increased on days 4-12, and thereafter the matrix was calcified on day 14. However, knockdown of MSX1 with small interfering RNA abolished the induction of the osteoblast-related gene expression, alkaline phosphatase activity, and calcification. Interestingly, DNA microarray and PCR analyses revealed that MSX1 knockdown induced the sterol regulatory element-binding protein 2 (SREBP2) transcriptional factor and its downstream target genes in the cholesterol synthesis pathway. Inhibition of cholesterol synthesis enhances osteoblast differentiation of various mesenchymal cells. Thus, MSX1 may downregulate the cholesterol synthesis-related genes to ensure osteoblast differentiation of human dental pulp stem cells.

5.
Int J Mol Med ; 38(3): 876-84, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27430159

ABSTRACT

Differentiated embryo chondrocyte 2 (DEC2) is a basic helix-loop-helix-Orange transcription factor that regulates cell differentiation in various mammalian tissues. DEC2 has been shown to suppress the differentiation of mesenchymal stem cells (MSCs) into myocytes and adipocytes. In the present study, we examined the role of DEC2 in the chondrogenic differentiation of human MSCs. The overexpression of DEC2 exerted minimal effects on the proliferation of MSCs in monolayer cultures with the growth medium under undifferentiating conditions, whereas it suppressed increases in DNA content, glycosaminoglycan content, and the expression of several chondrocyte-related genes, including aggrecan and type X collagen alpha 1, in MSC pellets in centrifuge tubes under chondrogenic conditions. In the pellets exposed to chondrogenesis induction medium, DEC2 overexpression downregulated the mRNA expression of fibroblast growth factor 18, which is involved in the proliferation and differentiation of chondrocytes, and upregulated the expression of p16INK4, which is a cell cycle inhibitor. These findings suggest that DEC2 is a negative regulator of the proliferation and differentiation of chondrocyte lineage-committed mesenchymal cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Chondrocytes/metabolism , Mesenchymal Stem Cells/metabolism , Aggrecans/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Lineage/genetics , Cells, Cultured , Chondrocytes/cytology , Collagen Type X/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA/genetics , DNA/metabolism , Extracellular Matrix/metabolism , Fibroblast Growth Factors/genetics , Gene Expression Regulation , Glycosaminoglycans/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mice , Reverse Transcriptase Polymerase Chain Reaction
6.
Curr Top Dev Biol ; 110: 339-72, 2014.
Article in English | MEDLINE | ID: mdl-25248482

ABSTRACT

Daily physiological and behavioral rhythms are regulated by endogenous circadian molecular clocks. Clock proteins DEC1 (BHLHe40) and DEC2 (BHLHe41) belong to the basic helix-loop-helix protein superfamily, which contains other clock proteins CLOCK and BMAL1. DEC1 and DEC2 are induced by CLOCK:BMAL1 heterodimer via the CACGTG E-box in the promoter and, thereafter, suppress their own expression by competing with CLOCK:BMAL1 for the DNA binding. This negative feedback DEC loop together with the PER loop involving PER and CRY, the other negative clock regulators, maintains the circadian rhythm of Dec1 and Dec2 expression. DEC1 is induced by light pulse and adjusts the circadian phase of the central clock in the suprachiasmatic nucleus, whereas DEC1 upregulation by TGF-ß resets the circadian phase of the peripheral clocks in tissues. Furthermore, DEC1 and DEC2 modulate the clock output signals to control circadian rhythms in behavior and metabolism. In addition to the functions in the clocks, DEC1 and DEC2 are involved in hypoxia responses, immunological reactions, and carcinogenesis. These DEC actions are mediated by the direct binding to the E-box elements in target genes or by protein-protein interactions with transcription factors such as HIF-1α, RXRα, MyoD, and STAT. Notably, numerous growth factors, hormones, and cytokines, along with ionizing radiation and DNA-damaging agents, induce Dec1 and/or Dec2 in a tissue-specific manner. These findings suggest that DEC1 and DEC2 play a critical role in animal adaptation to various environmental stimuli.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Circadian Rhythm/physiology , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , DNA/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Mice , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Structure-Activity Relationship , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
7.
J Biochem ; 154(4): 373-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23940085

ABSTRACT

We screened circadian-regulated genes in rat cartilage by using a DNA microarray analysis. In rib growth-plate cartilage, numerous genes showed statistically significant circadian mRNA expression under both 12:12 h light-dark and constant darkness conditions. Type II collagen and aggrecan genes--along with several genes essential for post-translational modifications of collagen and aggrecan, including prolyl 4-hydroxylase 1, lysyl oxidase, lysyl oxidase-like 2 and 3'-phosphoadenosine 5'-phosphosulphate synthase 2--showed the same circadian phase. In addition, the mRNA level of SOX9, a master transcription factor for the synthesis of type II collagen and aggrecan, has a similar phase of circadian rhythms. The circadian expression of the matrix-related genes may be critical in the development and the growth of various cartilages, because similar circadian expression of the matrix-related genes was observed in hip joint cartilage. However, the circadian phase of the major matrix-related genes in the rib permanent cartilage was almost the converse of that in the rib growth-plate cartilage under light-dark conditions. We also found that half of the oscillating genes had conserved clock-regulatory elements, indicating contribution of the elements to the clock outputs. These findings suggest that the synthesis of the cartilage matrix macromolecules is controlled by cell-autonomous clocks depending upon the in vivo location of cartilage.


Subject(s)
Cartilage/metabolism , Circadian Clocks , Matrilin Proteins/metabolism , Photoperiod , Animals , Gene Expression , Humans , Male , Matrilin Proteins/genetics , Rats , Rats, Sprague-Dawley
8.
Gene ; 510(2): 118-25, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22960268

ABSTRACT

Several cis-acting elements play critical roles in maintaining circadian expression of clock and clock-controlled genes. Using in silico analysis, we identified 10 sequence motifs that are correlated with the circadian phases of gene expression in the cartilage. One of these motifs, an E-box-like clock-related element (EL-box; GGCACGAGGC), can mediate BMAL1/CLOCK-induced transcription, which is typically regulated through an E-box or E'-box. Expression of EL-box-containing genes, including Ank, Dbp, and Nr1d1 (Rev-erbα), was induced by BMAL1/CLOCK or BMAL1/NPAS2. Compared with the E-box, the EL-box elements had distinct responsiveness to DEC1, DEC2, and HES1: suppressive actions of DEC1 and DEC2 on the EL-box were less potent than those on the E-box. HES1, which is known to bind to the N-box (CACNAG), suppressed enhancer activity of the EL-box, but not the E-box. In the Dbp promoter, an EL-box worked cooperatively with a noncanonical (NC) E-box to mediate BMAL1/CLOCK actions. These findings suggest that in addition to known clock elements, the EL-box element may contribute to circadian regulation of clock and clock-controlled genes.


Subject(s)
ARNTL Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins/metabolism , Cartilage/metabolism , Circadian Clocks , Homeodomain Proteins/metabolism , Animals , Base Sequence , Consensus Sequence , Growth Plate/metabolism , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Regulatory Elements, Transcriptional , Transcription Factor HES-1 , Transcriptome
9.
Int J Oncol ; 41(4): 1337-46, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22825629

ABSTRACT

DEC1 (BHLHE40/Stra13/Sharp2) is a basic helix-loop-helix (bHLH) transcription factor that is involved in the regulation of apoptosis and cell proliferation and the response to hypoxia. Epithelial-mesenchymal transition (EMT) is an important step leading to invasion and migration of various tumor cells, and TGF-ß treatment has been shown to induce cancer cells to undergo EMT. However, the significance of DEC1 in TGF-ß-induced EMT remains unknown. We examined the role of DEC1 in EMT of PANC-1 cells, a human pancreatic cancer cell line. As a result, we found that DEC1 was upregulated by TGF-ß in PANC-1 cells, and regulated the expression and the levels of nuclear, cytoplasmic or membrane localization of EMT-related factors, including phosphorylated Smad3 (pSmad3), snail, claudin-4 and N-cadherin. In the presence of TGF-ß, DEC1 knockdown by siRNA inhibited morphological changes during EMT processes, while TGF-ß induced PANC-1 cells to taken on a spindle-shaped morphology. Furthermore, a combination treatment of DEC1 expression with TGF-ß was closely linked to the migration and invasion of PANC-1 cells. Immunohistochemically, DEC1 and pSmad3 were detected within pancreatic cancer tissues, whereas claudin-4 expression was weaker in the cancer tissues compared with the adjacent non-cancer pancreatic tissues. These findings suggest that DEC1 plays an important role in the regulation of these EMT-related factors in pancreatic cancer.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Tumor Suppressor Proteins/genetics , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Claudin-4/metabolism , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/biosynthesis
10.
Int J Mol Med ; 30(3): 495-501, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22735690

ABSTRACT

The basic helix loop helix (bHLH) transcription factor DEC2 is associated with the regulation of apoptosis, circadian rhythm and the response to hypoxia. However, the significance of DEC2 in pancreatic cancer remains unknown. Here, we showed for the first time that DEC2 inhibits the progression of human pancreatic cancer. Human pancreatic cancer BxPC-3 cells were treated with or without transforming growth factor-ß (TGF-ß), siRNA against DEC2, or a combination of TGF-ß and DEC2 siRNA or DEC2 overexpression. The cells were analyzed by RT-PCR, real-time PCR, western blotting, immunofluorescent staining and ChIP assay. We also performed immunohistochemical analyses of DEC2 expression in surgically-resected pancreatic cancers. The expression of DEC2 was increased in TGF-ß-treated BxPC-3 cells. In the presence of TGF-ß, DEC2 overexpression decreased the migration and invasion of BxPC-3 cells. Knockdown of DEC2 by siRNA in the presence of TGF-ß significantly increased the expression and nuclear concentration of slug. We also showed that DEC2 siRNA decreased the binding of DEC2 to the E-box of the slug promoter. Immunohistochemically, little DEC2 was detected in pancreatic cancer tissues, whereas significant amounts were detected in the adjacent non-cancerous pancreatic tissues. These results indicate that DEC2 has inhibitory effects against human pancreatic cancer that involve TGF-ß and slug.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Pancreatic Neoplasms/metabolism , Transforming Growth Factor beta/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Pancreatic Neoplasms/genetics , RNA Interference , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Biomed Res ; 33(2): 75-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22572381

ABSTRACT

DEC1 (BHLHE40/Stra13/Sharp2) and DEC2 (BHLHE41/Sharp1) are basic helix-loop-helix (bHLH) transcription factors that are involved in the regulation of apoptosis, cell proliferation, circadian rhythms and the response to hypoxia. We previously showed the functional effects of DEC1 and DEC2 on apoptosis in human breast cancer MCF-7 cells. However, the roles of DEC1 and DEC2 in oral cancer are poorly understood. We examined whether DEC1 and DEC2 are involved in the regulation of apoptosis in human oral cancer HSC-3 and CA9-22 cells. The expression of DEC2 was upregulated by cis-diamminedichloroplatinum (II) (cisplatin: CDDP) treatment in HSC-3 cells, whereas CDDP treatment had little effects on the expression of DEC2 in CA9-22 cells. We showed that DEC2 overexpression inhibits pro-apoptotic factor Bim and inhibits apoptosis induced by CDDP in HSC-3 cells, whereas it had little effects on apoptosis in CA9-22 cells. DEC1 overexpression had little effects on apoptosis induced by CDDP in these cells. We also found that CDDP upregulated the amounts of DEC2 in the nucleus in HSC-3 cells. These results suggest that DEC2 has anti-apoptotic effects on apoptosis induced by CDDP in HSC-3 cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/physiology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bcl-2-Like Protein 11 , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cisplatin/pharmacology , Gene Expression/drug effects , Humans , Membrane Proteins/genetics , Mouth Neoplasms , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/physiology , Up-Regulation/drug effects
12.
Biochem Biophys Res Commun ; 419(2): 441-6, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22382019

ABSTRACT

Smads are intracellular signaling mediators. Complexes of Smad2 and Smad3 with Smad4 transmit transforming growth factor-beta (TGF-ß) receptor-induced signaling. Snail plays important roles in mesoderm formation, gastrulation, neural crest development, and epithelial mesenchymal transition. However, it remains unknown whether Smad3 and Snail expression is circadian rhythm-dependent. Here, we showed for the first time that Smad3 and Snail show circadian expression in human gingival fibroblasts (HGF-1) and human mesenchymal stem cells (MSC) after serum shock. They also showed circadian expression in the mouse liver. We confirmed that BMAL1/2, DEC1/2, VEGF, and PER1/2/3 also show circadian expression in both HGF-1 and MSC. The mRNA peaks and phases in circadian expression of these genes differed between HGF-1 and MSC. In a luciferase assay, Smad3 promoter activity was upregulated by CLOCK/BMAL1. These findings suggest that Smad3 and Snail have circadian rhythm in vitro and vivo, and that circadian expression of Smad3 depends on CLOCK/BMAL1.


Subject(s)
Circadian Rhythm , Fibroblasts/metabolism , Gingiva/metabolism , Liver/metabolism , Mesenchymal Stem Cells/metabolism , Smad3 Protein/biosynthesis , Transcription Factors/biosynthesis , ARNTL Transcription Factors/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , CLOCK Proteins/metabolism , Cells, Cultured , Humans , Male , Mice , Period Circadian Proteins/biosynthesis , Snail Family Transcription Factors , Tumor Suppressor Proteins/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
13.
Genes Cells ; 17(2): 109-21, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22244086

ABSTRACT

DEC1 and DEC2, members of the basic helix-loop-helix superfamily, are involved in various biological phenomena including clock systems, cell differentiation and metabolism. In clock systems, Dec1 and Dec2 expression are up-regulated by the CLOCK:BMAL1 heterodimer via E-box (CACGTG), exhibiting a circadian rhythm in the suprachiasmatic nucleus (SCN), the central circadian pacemaker and other peripheral tissues. In this study, using assays of luciferase reporters, electrophoretic mobility shift and chromatin immunoprecipitation, we identified novel nuclear receptor response elements, ROR response elements (RORE), in Dec1 and Dec2 promoters. These ROREs responded to the transcriptional activator RORα, but not to the repressor REVERBα, although the Bmal1 promoter responded to both RORα and REVERBα. Therefore, RORα, but not REVERBα, is involved in the regulation of Dec1 and Dec2 expression without significantly affecting their rhythmicity. Since RORα, DEC1 and DEC2 reportedly suppressed adipogenic differentiation, we examined expression of Rorα, Dec1, Dec2 and other clock-controlled genes in differentiating 3T3-L1 adipocytes. The results suggested that RORα suppresses adipogenic differentiation at a later stage of differentiation by RORE-mediated stimulation of Dec1 and Dec2 expression.


Subject(s)
Adipogenesis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation , Homeodomain Proteins/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Transcription Factors/genetics , ARNTL Transcription Factors/genetics , Animals , Base Sequence , Binding Sites , Cell Line , Circadian Rhythm/genetics , Gene Expression Profiling , Gene Order , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Response Elements
14.
Int J Mol Med ; 27(4): 491-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21327324

ABSTRACT

Differentiated embryonic chondrocyte gene (DEC) 1 (BHLHE40/Stra13/Sharp2) and DEC2 (BHLHE41/Sharp1) are basic helix-loop-helix (bHLH) transcription factors that are associated with the regulation of apoptosis, cell proliferation and circadian rhythms, as well as malignancy in various cancers. However, the roles of DEC1 and DEC2 expression in breast cancer are poorly understood. In this study, we sought to examine the roles of DEC1 and DEC2 in MCF-7 human breast cancer cells that had been treated with paclitaxel. The expression of DEC1 and DEC2 was up-regulated in paclitaxel-treated MCF-7 cells. Knockdown of DEC1 by siRNA decreased the amount of cleaved poly (ADP-ribose) polymerase (PARP), after treatment with paclitaxel, whereas DEC2 knockdown increased the amount of cleaved PARP in both the presence and absence of paclitaxel. Immunofluorescent staining revealed that paclitaxel treatment increased the amount of DEC1 in the nucleus, and increased the amount of DEC2 in both the nucleus and cytoplasm. These results indicate that DEC1 has pro-apoptotic effects, whereas DEC2 has anti-apoptotic effects on the paclitaxel-induced apoptosis in human breast cancer MCF-7 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Paclitaxel/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/physiopathology , Cell Line, Tumor , Cell Nucleus/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Protein Transport/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
15.
Genes Cells ; 15(4): 315-25, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20236182

ABSTRACT

DEC1 (BHLHB2/Stra13/Sharp2) and DEC2 (BHLHB3/Sharp1) are basic helix-loop-helix (bHLH) transcription factors that are involved in circadian rhythms, differentiation and the responses to hypoxia. We examined whether DEC1 and DEC2 are involved in apoptosis regulation, in human breast cancer MCF-7 cells. We found that siRNA-mediated knockdown of DEC2 resulted in marked enhancement of apoptosis compared with that in control cells transfected with nonspecific siRNA. However, knockdown of DEC1 by siRNA did not affect cell survival. Knockdown of DEC2 affected the expression of mRNA or proteins related to apoptosis, such as Fas, c-Myc, caspase-8, poly (ADP-ribose) polymerase (PARP) and Bax. We also showed that tumor necrosis factor-alpha (TNF-alpha) up-regulates the expression of DEC1 and DEC2. DEC2 over-expression caused by the transfection of an expression vector reduced the amounts of cleaved PARP and caspase-8 induced by TNF-alpha treatment, whereas DEC1 over-expression increased it. Finally, we revealed that treatment with double knockdown against both DEC1 and DEC2 decreased the amounts of cleaved PARP and caspase-8 induced by DEC2 siRNA with or without TNF-alpha. These data indicate that DEC2 has an anti-apoptotic effect, whereas DEC1 has a pro-apoptotic effect, which are involved in the balance of survival of human breast cancer MCF-7 cells.


Subject(s)
Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adenosine Diphosphate Ribose/genetics , Adenosine Diphosphate Ribose/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Breast Neoplasms/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Female , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factors/physiology , Transfection , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics
16.
Nihon Rinsho ; 67(8): 1454-7, 2009 Aug.
Article in Japanese | MEDLINE | ID: mdl-19768923
17.
Mol Pharmacol ; 76(6): 1360-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19786558

ABSTRACT

The basic helix-loop-helix proteins differentiated embryo chondrocyte 1 (DEC1) and DEC2 are involved in circadian rhythm control. Because the metabolism of dietary nutrients has been linked to circadian regulation, we examined the effect of DEC1 and DEC2 on the function of the metabolite-sensing nuclear receptors, ligand-dependent transcription factors, including retinoid X receptor (RXR) and liver X receptor (LXR). Transfection assays showed that DEC1 and DEC2 repressed ligand-dependent transactivation by RXR. Knockdown of endogenous DEC1 and DEC2 expression with small interfering RNAs augmented ligand-dependent RXRalpha transactivation. DEC1 and DEC2 interacted directly with RXRalpha, and ligand addition enhanced their association. DEC1 and DEC2 modified interaction of RXRalpha with cofactor proteins. Transfection assays using DEC1 and DEC2 mutants revealed that the C-terminal region of DEC2 is required for repression and that an LXXLL motif in DEC1 and DEC2 is necessary for RXRalpha repression. DEC1 and DEC2 repressed the induction of LXR target genes, associated with the promoter of an LXR target gene, and dissociated from the promoter with ligand treatment. Knockdown of endogenous DEC1 and DEC2 enhanced the LXR target gene expression in hepatocytes. Expression of Dec1, Dec2, and Srebp-1c showed a circadian rhythm in the liver of mice, whereas that of Lxralpha, Lxrbeta, and Rxralpha was not rhythmic. DEC1 and DEC2 also repressed the transactivation of other RXR heterodimers, such as farnesoid X receptor, vitamin D receptor, and retinoic acid receptor. Thus, the repressor function of DEC1 and DEC2 may be extended to other RXR heterodimer nuclear receptors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Chondrocytes/physiology , Retinoid X Receptors/physiology , Tumor Suppressor Proteins/physiology , Animals , Cell Line , Down-Regulation , Glutathione Transferase/biosynthesis , Histone Deacetylases/physiology , Homeodomain Proteins/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Retinoid X Receptor alpha/biosynthesis , Retinoid X Receptor alpha/physiology , Retinoid X Receptors/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/physiology , Transcriptional Activation/physiology , Transfection
18.
Genes Cells ; 14(1): 29-40, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19032342

ABSTRACT

DEC1 (BHLHB2/Stra13/Sharp2)-a basic helix-loop-helix transcription factor-is known to be involved in various biological phenomena including clock systems and metabolism. In the clock systems, Dec1 expression is dominantly up-regulated by CLOCK : BMAL1 heterodimer, and it exhibits circadian rhythm in the suprachiasmatic nucleus (SCN)-the central circadian pacemaker-and other peripheral tissues. Recent studies have shown that the strong circadian rhythmicity of Dec1 in the SCN was abolished by Clock mutation, whereas that in the liver was affected, but not abolished, by Clock mutation. Moreover, feeding conditions affected hepatic Dec1 expression, which indicates that Dec1 expression is closely linked with the metabolic functions of the liver. Among ligand-activated nuclear receptors examined, LXRalpha and LXRbeta with T0901317-agonist for LXR-were found to be potent enhancers for Dec1 promoter activity, and a higher expression level of LXRalpha protein was detected in the liver than in the kidney and heart. T0901317 increased the levels of endogenous Dec1 transcript in hepatoma cells. Chromatin immunoprecipitation assay indicated that LXRalpha bound to the Dec1 promoter, and an LXRalpha-binding site was identified. These observations indicate that hepatic DEC1 mediates the ligand-dependent LXR signal to regulate the expression of genes involved in the hepatic clock system and metabolism.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Binding Sites , CLOCK Proteins , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , Dimerization , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Liver X Receptors , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NIH 3T3 Cells , Organ Specificity/genetics , Orphan Nuclear Receptors , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Response Elements/genetics , Trans-Activators/metabolism
19.
Mol Cell Biol ; 28(12): 4080-92, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18411297

ABSTRACT

DEC1 suppresses CLOCK/BMAL1-enhanced promoter activity, but its role in the circadian system of mammals remains unclear. Here we examined the effect of Dec1 overexpression or deficiency on circadian gene expression triggered with 50% serum. Overexpression of Dec1 delayed the phase of clock genes such as Dec1, Dec2, Per1, and Dbp that contain E boxes in their regulatory regions, whereas it had little effect on the circadian phase of Per2 and Cry1 carrying CACGTT E' boxes. In contrast, Dec1 deficiency advanced the phase of the E-box-containing clock genes but not that of the E'-box-containing clock genes. Accordingly, DEC1 showed strong binding and transrepression on the E box, but not on the E' box, in chromatin immunoprecipitation, electrophoretic mobility shift, and luciferase reporter assays. Dec1-/- mice showed behavioral rhythms with slightly but significantly longer circadian periods under conditions of constant darkness and faster reentrainment to a 6-h phase-advanced shift of a light-dark cycle. Knockdown of Dec2 with small interfering RNA advanced the phase of Dec1 and Dbp expression, and double knockdown of Dec1 and Dec2 had much stronger effects on the expression of the E-box-containing clock genes. These findings suggest that DEC1, along with DEC2, plays a role in the finer regulation and robustness of the molecular clock.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Trans-Activators/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Circadian Rhythm , Female , Humans , Male , Mice , Mice, Transgenic , NIH 3T3 Cells , Stem Cells/cytology
20.
Genes Cells ; 13(2): 131-44, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18233956

ABSTRACT

DEC1 (BHLHB2/Sharp2/Stra13) and DEC2 (BHLHB3/Sharp1) are basic-helix-loop-helix (bHLH) transcription factors, involved in cellular differentiation, responses to hypoxia and circadian rhythms. We recently showed that the expression of DEC1 and DEC2 was up-regulated by hypoxia; however, the functions of these two factors under hypoxic conditions have not been elucidated in detail. It is well established that the expression of vascular endothelial growth factor (VEGF) is up-regulated by hypoxia, and the expression of VEGF in response to hypoxia depends on transcriptional activation by a heterodimer comprising hypoxia-inducible factor 1alpha (HIF-1alpha) and arylhydrocarbon receptor nuclear translocator 1 (ARNT1). In the present study, we showed that DEC2, but not DEC1, suppressed VEGF gene expression under hypoxic conditions. DEC2 protein was co-immunoprecipitated with HIF-1alpha but not with ARNT1. The binding of HIF-1alpha to the hypoxia response element (HRE) in the VEGF promoter was decreased by DEC2 over-expression, and increased by DEC2 knockdown. We also showed that the circadian expression of VEGF showed a reciprocal pattern to that of DEC2 in cartilage. DEC2 had a circadian oscillation in implanted Sarcoma 180 cells. We conclude that DEC2 negatively regulates VEGF expression and plays an important role in the pathological conditions in which VEGF is involved.


Subject(s)
Cell Hypoxia/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Plasmids/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcoma 180/genetics , Sarcoma 180/metabolism , Transcription Factors/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...