Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 125(18): 4831-4837, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33891418

ABSTRACT

Triplet-triplet annihilation (TTA)-assisted photon upconversion (TTA-UC) in three dyads (DPA-Cn-DPA), comprised of two diphenylanthracene (DPA) moieties connected by nonconjugated C1, C2, and C3 linkages (Cn), has been investigated. The performance of these dyads as energy acceptors in the presence of the energy donor platinum octaethylporphyrin are characterized by longer triplet lifetimes (τT) and different TTA rate constants than those of the parent DPA. The larger τT of the linked systems, caused by "intramolecular energy hopping" in the triplet dyad 3DPA*-Cn-DPA, results in a low threshold intensity, a key characteristic of efficient TTA-UC.

2.
Stem Cell Res Ther ; 12(1): 203, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33757592

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) provide potential treatments for peritoneal fibrosis. However, MSCs cultured in media containing serum bring risks of infection and other problems. In this study, we compared the effect of human MSCs in serum-free medium (SF-MSCs) on peritoneal fibrosis with that of MSCs cultured in medium containing 10% fetal bovine serum (10%MSCs). METHODS: Peritoneal fibrosis was induced by intraperitoneally injecting 0.1% chlorhexidine gluconate (CG). SF-MSCs or 10%MSCs were intraperitoneally administered 30 min after the CG injection. Ten days after the CG and MSC injections, we performed histological analyses and peritoneal equilibrium testing. In the in vitro experiments, we used transforming growth factor (TGF)-ß1-stimulated human peritoneal mesothelial cells incubated in conditioned medium from MSCs to examine whether the SF-MSCs showed enhanced ability to produce antifibrotic humoral factors. RESULTS: Histological staining showed that the SF-MSCs significantly suppressed CG-induced cell accumulation and thickening compared with that of the 10%MSCs. Additionally, the SF-MSCs significantly inhibited mesenchymal cell expression, extracellular matrix protein deposition and inflammatory cell infiltration. Peritoneal equilibration testing showed that compared with administering 10%MSCs, administering SF-MSCs significantly reduced the functional impairments of the peritoneal membrane. The in vitro experiments showed that although the conditioned medium from MSCs suppressed TGF-ß1 signaling, the suppression did not significantly differ between the SF-MSCs and 10%MSCs. CONCLUSIONS: Serum-free culture conditions can enhance the antifibrotic abilities of MSCs by suppressing inflammation. Administering ex vivo expanded SF-MSCs may be a potential therapy for preventing peritoneal fibrotic progression.


Subject(s)
Mesenchymal Stem Cells , Peritoneal Fibrosis , Cells, Cultured , Culture Media, Conditioned/pharmacology , Humans , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/genetics , Peritoneal Fibrosis/therapy , Peritoneum , Serum
3.
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
4.
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
5.
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.

6.
Clin Exp Nephrol ; 20(1): 30-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26018137

ABSTRACT

BACKGROUND: High glucose (HG) induces production of transforming growth factor-beta1 (TGF-ß1), but the mechanism remains elusive. The aim of this study was to determine the gene(s) involved in HG-induced TGF-ß1 production in human peritoneal mesothelial cells (HPMCs). METHODS: Microarray analysis was performed following a 3-h preincubation of HPMCs in 4 or 0.1% glucose medium. Transcriptional genes were selected using Gene Ontology analysis for biological processes, including regulation of transcription and DNA-dependent. The effects of small interfering RNA (siRNA) treatments on the up-regulation of TGF-ß1 mRNA were assessed by quantitative real-time polymerase chain reaction (qPCR). Finally, enzyme-linked immunosorbent assay (ELISA) was performed to determine which gene(s) contribute to the production of TGF-ß1 protein in the medium. RESULTS: Microarray analysis revealed that the expression of 51 genes increased by more than 3-fold. Gene ontology analysis identified 13 genes for further study. qPCR confirmed mRNA amplification for 9 of the 13 genes. Furthermore, HG-induced up-regulation of TGF-ß1 mRNA was attenuated by the siRNA of 4 genes: MDS1 and EVI1 complex locus (MECOM), FBJ murine osteosarcoma viral oncogene homolog B (FOSB), FBJ murine osteosarcoma viral oncogene homolog (FOS) and activating transcription factor 3 (ATF3). ELISA showed that siRNA treatment of FOS, but not MECOM, FOSB or ATF3, suppressed the increase of TGF-ß1 protein in the medium. CONCLUSIONS: FOS is a downstream effector of HG stimulation in HPMCs that contributes to TGF-ß1 production, suggesting that blocking FOS expression may be a therapeutic target for peritoneal fibrosis.


Subject(s)
Glucose/pharmacology , Peritoneum/drug effects , Peritoneum/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Peritoneum/cytology , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic , Transfection , Transforming Growth Factor beta1/genetics , Up-Regulation
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.
Kidney Int ; 84(2): 297-307, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23486522

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent adult stem cells that have regenerative capability and exert paracrine actions on damaged tissues. Since peritoneal fibrosis is a serious complication of peritoneal dialysis, we tested whether MSCs suppress this using a chlorhexidine gluconate model in rats. Although MSCs isolated from green fluorescent protein-positive rats were detected for only 3 days following their injection, immunohistochemical staining showed that MSCs suppressed the expression of mesenchymal cells, their effects on the deposition of extracellular matrix proteins, and the infiltration of macrophages for 14 days. Moreover, MSCs reduced the functional impairment of the peritoneal membrane. Cocultures of MSCs and human peritoneal mesothelial cells using a Transwell system indicated that the beneficial effects of MSCs on the glucose-induced upregulation of transforming growth factor-ß1(TGF-ß1) and fibronectin mRNA expression in the human cells were likely due to paracrine actions. Preincubation in MSC-conditioned medium suppressed TGF-ß1-induced epithelial-to-mesenchymal transition, α-smooth muscle actin, and the decrease in zonula occludens-1 in cultured human peritoneal mesothelial cells. Although bone morphogenic protein 7 was not detected, MSCs secreted hepatocyte growth factor and a neutralizing antibody to this inhibited TGF-ß1 signaling. Thus, our findings imply that MSCs ameliorate experimental peritoneal fibrosis by suppressing inflammation and TGF-ß1 signaling in a paracrine manner.


Subject(s)
Inflammation Mediators/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Peritoneal Fibrosis/prevention & control , Peritoneum/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Chemotaxis , Chlorhexidine/analogs & derivatives , Coculture Techniques , Culture Media, Conditioned/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Extracellular Matrix Proteins/metabolism , Glucose/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Male , Mesenchymal Stem Cells/immunology , Paracrine Communication , Peritoneal Fibrosis/chemically induced , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneum/immunology , Peritoneum/pathology , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Smad2 Protein/metabolism , Time Factors
9.
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
10.
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
11.
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
12.
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
13.
J Biochem ; 140(3): 401-8, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16873396

ABSTRACT

To elucidate the food-entrainable oscillatory mechanism of peripheral clock systems, we examined the effect of fasting on circadian expression of clock genes including Dec1 and Dec2 in mice. Withholding of food for 2 days had these effects: the expression level of Dec1 mRNA decreased in all tissues examined, although Per1 mRNA level markedly increased; Per2 expression was reduced in the liver and heart only 42-46 h after the start of fasting; and expression profiles of Dec2 and Bmal1 were altered only in the heart and in the liver, respectively, whereas Rev-erbalpha mRNA levels did not change significantly. Re-feeding after 36-h starvation erased, at least in part, the effect of fasting on Dec1, Dec2, Per1, Per2, and Bmal1 within several hours, and restriction feeding shifted the phase of expression profiles of all examined clock genes including Dec1 and Dec2. These findings indicate that short-term fasting and re-feeding modulate the circadian rhythms of clock genes to different extents in peripheral tissues, and suggest that the expression of Dec1, Per1, and some other clock genes was closely linked with the metabolic activity of these tissues.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Biological Clocks/physiology , Cell Cycle Proteins/metabolism , Circadian Rhythm/physiology , Fasting/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Period Circadian Proteins , Time Factors
14.
Clin Calcium ; 16(5): 838-45, 2006 May.
Article in Japanese | MEDLINE | ID: mdl-16679627

ABSTRACT

Circadian rhythms in cartilage have been reported repeatedly. However, previous studies used histological analysis or radioisotope-labeled precursors for DNA, collagen and proteoglycan synthesis, and thus it is difficult precisely to evaluate such studies on circadian rhythms in chondrocytes. On the other hand, circadian rhythms in plasma levels of several hormones, which play crucial roles in cartilage metabolism, proved to be significant both in human and animal models. In addition, clock genes--such as Clock, Bmal, Per, Cry and Dec--were identified in suprachiasmatic nucleus (SCN) and some peripheral tissues. These clock genes may be involved in circadian rhythms in cartilage.


Subject(s)
Cartilage/metabolism , Circadian Rhythm/genetics , Circadian Rhythm/physiology , Animals , CLOCK Proteins , Cell Division/genetics , Chondrocytes/cytology , Collagen/biosynthesis , DNA/biosynthesis , Humans , Proteoglycans/biosynthesis , Trans-Activators/physiology
15.
Eur J Biochem ; 271(22): 4409-19, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15560782

ABSTRACT

The basic helix-loop-helix transcription factor DEC1 is expressed in a circadian manner in the suprachiasmatic nucleus where it seems to play a role in regulating the mammalian circadian rhythm by suppressing the CLOCK/BMAL1-activated promoter. The interaction of DEC1 with BMAL1 has been suggested as one of the molecular mechanisms of the suppression [Honma, S., Kawamoto, T., Takagi, Y., Fujimoto, K., Sato, F., Noshiro, M., Kato, Y. & Honma, K. (2002) Nature 419, 841-844]. Deletion analysis of DEC1 demonstrated that its N-terminal region, which includes the basic helix-loop-helix domain, was essential for both the suppressive activity and the interaction with BMAL1, as DEC1 lacking the basic region did not show any suppression or interaction. Furthermore, we found that Arg65 in the basic region, which is conserved among group B basic helix-loop-helix proteins, was responsible for the suppression, for the interaction with BMAL1 and for its binding to CACGTG E-boxes. However, substitution of His57 for Ala significantly reduced the E-box binding activity of DEC1, although it did not affect the interaction with BMAL1 or suppression of CLOCK/BMAL1-induced transcription. On the other hand, the basic region-deleted DEC1 acted in a dominant-negative manner for DEC1 activity, indicating that the basic region was not required for homodimer formation of DEC1. Moreover, mutant DEC1 also counteracted DEC2-mediated suppressive activity in a dominant-negative manner. The heterodimer formation of DEC1 and DEC2 was confirmed by pull-down assay. These findings suggest that the basic region of DEC1 participates in the transcriptional regulation through a protein-protein interaction with BMAL1 and DNA binding to the E-box.


Subject(s)
Circadian Rhythm/physiology , Homeodomain Proteins/physiology , Transcription Factors/metabolism , ARNTL Transcription Factors , Amino Acid Substitution , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , CLOCK Proteins , Circadian Rhythm/genetics , Dimerization , E-Box Elements/physiology , Gene Deletion , Genes, Dominant , Helix-Loop-Helix Motifs , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Humans , Luciferases/metabolism , Mice , NIH 3T3 Cells , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/physiology , Trans-Activators/physiology , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...