Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Atherosclerosis ; 229(1): 182-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23639859

ABSTRACT

Evaluation of atherosclerotic plaques depends on invasive intravascular ultrasonography (IVUS). Carboxy-terminal telopeptide of type I collagen (ICTP) is produced by matrix metalloproteinase (MMP)-dependent digestion of type I collagen. Because vulnerable plaques are rich in type I collagen and MMPs from macrophages, we examined the association between serum ICTP and coronary plaques in patients with coronary disease. We recruited 46 men and 17 women without renal failure or bone diseases affecting serum ICTP, who underwent coronary IVUS. Serum ICTP levels were higher in patients with coronary plaques containing more than 10% necrotic core area than in patients with less than 10% necrotic core area. A positive correlation was found between serum ICTP and necrotic core area. Only serum ICTP was positively correlated with necrotic core area by multivariate analysis (p < 0.05). These results suggest that serum ICTP can be used as a non-invasive marker of vulnerable plaques in atherosclerotic patients.


Subject(s)
Collagen Type I/blood , Coronary Artery Disease/blood , Coronary Artery Disease/epidemiology , Peptides/blood , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/epidemiology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/epidemiology , Acute Coronary Syndrome/pathology , Aged , Aged, 80 and over , Biomarkers/blood , Coronary Artery Disease/pathology , Cross-Sectional Studies , Female , Humans , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Morbidity , Necrosis , Pilot Projects , Plaque, Atherosclerotic/pathology , Risk Factors
2.
Endocrinology ; 154(3): 1156-67, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23397032

ABSTRACT

Glucocorticoid (GC) excess causes a rapid loss of bone with a reduction in bone formation. Intermittent PTH (1-34) administration stimulates bone formation and counteracts the inhibition of bone formation by GC excess. We have previously demonstrated that mechanical strain enhances interleukin (IL)-11 gene transcription by a rapid induction of ΔFosB expression and protein kinase C (PKC)-δ-mediated phosphorylation of phosphorylated mothers against decapentaplegic (Smad)-1. Because IL-11 suppresses the expression of dickkopf-1 and -2 and stimulates Wnt signaling, IL-11 appears to mediate at least a part of the effect of mechanical strain on osteoblast differentiation and bone formation. The present study was undertaken to examine the effect of PTH(1-34) and GCs on IL-11 expression in murine primary osteoblasts (mPOBs). PTH(1-34) treatment of mPOBs enhanced IL-11 expression in a time- and dose-dependent manner. PTH(1-34) also stimulated ΔFosB expression and Smad1 phosphorylation, which cooperatively stimulated IL-11 gene transcription. PTH(1-34)-induced Smad1 phosphorylation was mediated via PKCδ and was abrogated in mPOBs from PKCδ knockout mice. Dexamethasone suppressed IL-11 gene transcription enhanced by PTH(1-34) without affecting ΔFosB expression or Smad1 phosphorylation, and dexamethasone-GC receptor complex was bound to JunD, which forms heterodimers with ΔFosB. High doses of PTH(1-34) counteracted the effect of dexamethasone on apoptosis of mPOBs, which was blunted by neutralizing anti-IL-11 antibody or IL-11 small interfering RNA. These results demonstrate that PTH(1-34) and GCs interact to regulate IL-11 expression in parallel with osteoblast differentiation and apoptosis and suggest that PTH(1-34) and dexamethasone may regulate osteoblast differentiation and apoptosis via their effect on IL-11 expression.


Subject(s)
Interleukin-11/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Parathyroid Hormone/pharmacology , Alkaline Phosphatase/genetics , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Mice , Osteoblasts/cytology , Osteocalcin/genetics , Osteoprotegerin/genetics , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RANK Ligand/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Smad1 Protein/metabolism
3.
Endocr J ; 59(2): 91-101, 2012.
Article in English | MEDLINE | ID: mdl-21931225

ABSTRACT

Mechanical stress and parathyroid hormone (PTH) are major stimulators, and aging and glucocorticoids excess are important suppressors of osteoblast differentiation. Mechanical stress and PTH stimulate interleukin (IL)-11 expression in cells of osteoblast lineage by enhancing transcription of IL-11 gene via an increase in intracellular Ca²âº. The elevated Ca²âº activates extracellular signal-regulated kinase (ERK) to enhance phosphorylation of cyclic AMP response element-binding protein (CREB), which binds to the fosB gene promoter and enhances ΔFosB expression. ΔFosB dimerizes with JunD on the IL-11 gene promoter to enhance its transcription. Both mechanical stress and PTH also stimulate phosphorylation of Smad1 via an activation of protein kinase Cδ (PKCδ). Phosphorylated Smad1 binds to the IL-11 gene promoter and forms complex with ΔFosB/JunD to further enhance IL-11 gene transcription. The increased IL-11 then suppresses expression of Wnt inhibitors, including Dickkopf 1 (Dkk1) and 2, and enhances Wnt signaling to stimulate osteoblast differentiation and inhibit adipocyte differentiation. The suppression of osteoblast differentiation by aging involves a decrease in IL-11 gene transcription by a reduction in JunD binding to the activator protein (AP)-1 site of the IL-11 gene promoter. Glucocorticoids inhibit transcriptional activation of IL-11 gene by an interaction of glucocorticoid-glucocorticoid receptor (GR) complex with ΔFosB/JunD heterodimer. Thus, factors that enhance osteoblast differentiation stimulate, and those which suppress osteoblast differentiation inhibit IL-11 gene transcription, and IL-11 enhances Wnt signaling by suppressing expression of its inhibitors. These observations are consistent with the notion that IL-11 mediates stimulatory and inhibitory signals of osteoblast differentiation by affecting Wnt signaling.


Subject(s)
Interleukin-11/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Signal Transduction , Smad Proteins/metabolism , Transcription Factor AP-1/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Aging , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Humans , Wnt Signaling Pathway
4.
PLoS One ; 5(9)2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20927330

ABSTRACT

BACKGROUND: Mechanical stress rapidly induces ΔFosB expression in osteoblasts, which binds to interleukin (IL)-11 gene promoter to enhance IL-11 expression, and IL-11 enhances osteoblast differentiation. Because bone morphogenetic proteins (BMPs) also stimulate IL-11 expression in osteoblasts, there is a possibility that BMP-Smad signaling is involved in the enhancement of osteoblast differentiation by mechanical stress. The present study was undertaken to clarify whether mechanical stress affects BMP-Smad signaling, and if so, to elucidate the role of Smad signaling in mechanical stress-induced enhancement of IL-11 gene transcription. METHODOLOGY/PRINCIPAL FINDINGS: Mechanical loading by fluid shear stress (FSS) induced phosphorylation of BMP-specific receptor-regulated Smads (BR-Smads), Smad1/5, in murine primary osteoblasts (mPOBs). FSS rapidly phosphorylated Y311 of protein kinase C (PKC)δ, and phosphorylated PKCδ interacted with BR-Smads to phosphorylate BR-Smads. Transfection of PKCδ siRNA or Y311F mutant PKCδ abrogated BR-Smads phosphorylation and suppressed IL-11 gene transcription enhanced by FSS. Activated BR-Smads bound to the Smad-binding element (SBE) of IL-11 gene promoter and formed complex with ΔFosB/JunD heterodimer via binding to the C-terminal region of JunD. Site-directed mutagenesis in the SBE and the AP-1 site revealed that both SBE and AP-1 sites were required for full activation of IL-11 gene promoter by FSS. CONCLUSIONS/SIGNIFICANCE: These results demonstrate that PKCδ-BR-Smads pathway plays an important role in the intracellular signaling in response to mechanical stress, and that a cross-talk between PKCδ-BR-Smads and ΔFosB/JunD pathways synergistically stimulates IL-11 gene transcription in response to mechanical stress.


Subject(s)
Interleukin-11/genetics , Osteoblasts/chemistry , Osteoblasts/metabolism , Protein Kinase C-delta/metabolism , Signal Transduction , Smad Proteins/metabolism , Transcription, Genetic , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Line , Cells, Cultured , Humans , Interleukin-11/metabolism , Mice , Osteoblasts/enzymology , Phosphorylation , Protein Binding , Protein Kinase C-delta/genetics , Smad Proteins/genetics , Stress, Mechanical
5.
Bone ; 45(6): 1125-32, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19665600

ABSTRACT

Molecular mechanism of mechanical stress-induced bone formation remains unclear. We demonstrate that mechanical unloading suppresses and reloading enhances Interleukin (IL)-11 gene expression in the hindlimb of mice in vivo. Mechanical stress to osteoblasts by fluid shear stress (FSS) in vitro rapidly and transiently enhances fosB gene transcription, stimulates binding of DeltaFosB/JunD complex to activator protein (AP)-1 site of the IL-11 gene promoter, and enhances IL-11 gene transcription. Anti-IL-11 antibody blocks mechanical stress-induced enhancement of osteoblastogenesis and suppression of adipogenesis, suggesting the requirement of IL-11 for the stimulation of osteoblast differentiation by mechanical stress. Down-regulation of DeltaFosB/JunD by small interfering RNA (siRNA) suppresses and overexpression of DeltaFosB/JunD enhances IL-11 gene promoter activity. Consistent with our previous observations that up-regulation of DeltaFosB depends upon activation of cyclic AMP response element-binding protein (CREB) via Ca(2+)-dependent activation of extracellular signal-regulated kinase (ERK) to phosphorylate CREB, mechanical stress-induced activation of IL-11 gene transcription is dependent upon Ca(2+)-ERK pathway. Present results also demonstrated that FSS to osteoblasts enhances canonical Wnt signaling in vitro, and that mechanical unloading induces and reloading suppresses the expression of a canonical Wnt signal inhibitor, dickkopf2 (Dkk2), in vivo. In addition, IL-11 siRNA enhances Dkk2 expression suppressed by FSS, and osteoblasts from IL-11 transgenic mice show reduced Dkk2 mRNA expression than those from wild-type mice. These observations are consistent with the notion that mechanical stress stimulates IL-11 gene transcription via an enhanced DeltaFosB/JunD binding to the IL-11 gene promoter, and that increased IL-11 enhances canonical Wnt signal at least in part via a reduction in Dkk2 expression to stimulate osteoblast differentiation.


Subject(s)
Cell Differentiation/genetics , Gene Expression Regulation , Interleukin-11/genetics , Osteoblasts/metabolism , Stress, Mechanical , Adipogenesis , Animals , Calcium/metabolism , Down-Regulation/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-11/metabolism , MAP Kinase Signaling System , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteogenesis , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun , Rheology , Transcription Factor AP-1/metabolism , Transcription, Genetic , Weight-Bearing , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...