Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Diabetes ; 65(9): 2502-15, 2016 09.
Article En | MEDLINE | ID: mdl-27284105

Identification of new adipokines that potentially link obesity to insulin resistance represents a major challenge. We recently showed that NOV/CCN3, a multifunctional matricellular protein, is synthesized and secreted by adipose tissue, with plasma levels highly correlated with BMI. NOV involvement in tissue repair, fibrotic and inflammatory diseases, and cancer has been previously reported. However, its role in energy homeostasis remains unknown. We investigated the metabolic phenotype of NOV(-/-) mice fed a standard or high-fat diet (HFD). Strikingly, the weight of NOV(-/-) mice was markedly lower than that of wild-type mice but only on an HFD. This was related to a significant decrease in fat mass associated with an increased proportion of smaller adipocytes and to a higher expression of genes involved in energy expenditure. NOV(-/-) mice fed an HFD displayed improved glucose tolerance and insulin sensitivity. Interestingly, the absence of NOV was associated with a change in macrophages profile (M1-like to M2-like), in a marked decrease in adipose tissue expression of several proinflammatory cytokines and chemokines, and in enhanced insulin signaling. Conversely, NOV treatment of adipocytes increased chemokine expression. Altogether, these results show that NOV is a new adipocytokine that could be involved in obesity-associated insulin-resistance.


Adipose Tissue/metabolism , Nephroblastoma Overexpressed Protein/metabolism , Obesity/metabolism , 3T3-L1 Cells , Adipose Tissue/physiopathology , Animals , Body Composition/genetics , Body Composition/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Energy Metabolism/physiology , Female , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Insulin Resistance/physiology , Liver/metabolism , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nephroblastoma Overexpressed Protein/genetics , Obesity/physiopathology , Pancreas/metabolism , RNA, Small Interfering/genetics
2.
PLoS One ; 10(9): e0137876, 2015.
Article En | MEDLINE | ID: mdl-26367310

The main hallmark of chronic kidney disease (CKD) is excessive inflammation leading to interstitial tissue fibrosis. It has been recently reported that NOV/CCN3 could be involved in kidney damage but its role in the progression of nephropathies is poorly known. NOV/CCN3 is a secreted multifunctional protein belonging to the CCN family involved in different physiological and pathological processes such as angiogenesis, inflammation and cancers. The purpose of our study was to determine the role of NOV/CCN3 in renal inflammation and fibrosis related to primitive tubulointerstitial injury. After unilateral ureteral obstruction (UUO), renal histology and real-time PCR were performed in NOV/CCN3-/- and wild type mice. NOV/CCN3 mRNA expression was increased in the obstructed kidneys in the early stages of the obstructive nephropathy. Interestingly, plasmatic levels of NOV/CCN3 were strongly induced after 7 days of UUO and the injection of recombinant NOV/CCN3 protein in healthy mice significantly increased CCL2 mRNA levels. Furthermore, after 7 days of UUO NOV/CCN3-/- mice displayed reduced proinflammatory cytokines and adhesion markers expression leading to restricted accumulation of interstitial monocytes, in comparison with their wild type littermates. Consequently, in NOV/CCN3-/- mice interstitial renal fibrosis was blunted after 15 days of UUO. In agreement with our experimental data, NOV/CCN3 expression was highly increased in biopsies of patients with tubulointerstitial nephritis. Thus, the inhibition of NOV/CCN3 may represent a novel target for the progression of renal diseases.


Kidney Diseases/pathology , Kidney/pathology , Nephritis, Interstitial/pathology , Nephroblastoma Overexpressed Protein/metabolism , Animals , Biomarkers/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Gene Expression Regulation , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Nephritis, Interstitial/genetics , Nephritis, Interstitial/metabolism , Nephroblastoma Overexpressed Protein/blood , Nephroblastoma Overexpressed Protein/genetics , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/metabolism
3.
PLoS One ; 8(6): e66788, 2013.
Article En | MEDLINE | ID: mdl-23785511

OBJECTIVE: Evidence points to a founder of the multifunctional CCN family, NOV/CCN3, as a circulating molecule involved in cardiac development, vascular homeostasis and inflammation. No data are available on the relationship between plasma NOV/CCN3 levels and cardiovascular risk factors in humans. This study investigated the possible relationship between plasma NOV levels and cardiovascular risk factors in humans. METHODS: NOV levels were measured in the plasma from 594 adults with a hyperlipidemia history and/or with lipid-lowering therapy and/or a body mass index (BMI) >30 kg/m(2). Correlations were measured between NOV plasma levels and various parameters, including BMI, fat mass, and plasma triglycerides, cholesterol, glucose, and C-reactive protein. NOV expression was also evaluated in adipose tissue from obese patients and rodents and in primary cultures of adipocytes and macrophages. RESULTS: After full multivariate adjustment, we detected a strong positive correlation between plasma NOV and BMI (r = 0.36 p<0.0001) and fat mass (r = 0.33 p<0.0005). According to quintiles, this relationship appeared to be linear. NOV levels were also positively correlated with C-reactive protein but not with total cholesterol, LDL-C or blood glucose. In patients with drastic weight loss induced by Roux-en-Y bariatric surgery, circulating NOV levels decreased by 28% (p<0.02) and 48% (p<0.0001) after 3 and 6 months, respectively, following surgery. In adipose tissue from obese patients, and in human primary cultures NOV protein was detected in adipocytes and macrophages. In mice fed a high fat diet NOV plasma levels and its expression in adipose tissue were also significantly increased compared to controls fed a standard diet. CONCLUSION: Our results strongly suggest that in obese humans and mice plasma NOV levels positively correlated with NOV expression in adipose tissue, and support a possible contribution of NOV to obesity-related inflammation.


Metabolic Diseases/blood , Metabolic Diseases/complications , Nephroblastoma Overexpressed Protein/blood , Obesity/blood , Obesity/complications , Adipose Tissue/metabolism , Adult , Aged , Animals , Blood Glucose , Body Composition , Body Mass Index , C-Reactive Protein/metabolism , Diet, High-Fat , Female , Humans , Lipid Metabolism , Male , Metabolic Diseases/metabolism , Mice , Middle Aged , Nephroblastoma Overexpressed Protein/metabolism , Obesity/metabolism , Risk Factors
4.
J Neuroinflammation ; 9: 36, 2012 Feb 21.
Article En | MEDLINE | ID: mdl-22353423

BACKGROUND: Sustained neuroinflammation strongly contributes to the pathogenesis of pain. The clinical challenge of chronic pain relief led to the identification of molecules such as cytokines, chemokines and more recently matrix metalloproteinases (MMPs) as putative therapeutic targets. Evidence points to a founder member of the matricial CCN family, NOV/CCN3, as a modulator of these inflammatory mediators. We thus investigated the possible involvement of NOV in a preclinical model of persistent inflammatory pain. METHODS: We used the complete Freund's adjuvant (CFA)-induced model of persistent inflammatory pain and cultured primary sensory neurons for in vitro experiments. The mRNA expression of NOV and pro-inflammatory factors were measured with real-time quantitative PCR, CCL2 protein expression was assessed using ELISA, MMP-2 and -9 activities using zymography. The effect of drugs on tactile allodynia was evaluated by the von Frey test. RESULTS: NOV was expressed in neurons of both dorsal root ganglia (DRG) and dorsal horn of the spinal cord (DHSC). After intraplantar CFA injection, NOV levels were transiently and persistently down-regulated in the DRG and DHSC, respectively, occurring at the maintenance phase of pain (15 days). NOV-reduced expression was restored after treatment of CFA rats with dexamethasone. In vitro, results based on cultured DRG neurons showed that siRNA-mediated inhibition of NOV enhanced IL-1ß- and TNF-α-induced MMP-2, MMP-9 and CCL2 expression whereas NOV addition inhibited TNF-α-induced MMP-9 expression through ß1 integrin engagement. In vivo, the intrathecal delivery of MMP-9 inhibitor attenuated mechanical allodynia of CFA rats. Importantly, intrathecal administration of NOV siRNA specifically led to an up-regulation of MMP-9 in the DRG and MMP-2 in the DHSC concomitant with increased mechanical allodynia. Finally, NOV intrathecal treatment specifically abolished the induction of MMP-9 in the DRG and, MMP-9 and MMP-2 in the DHSC of CFA rats. This inhibitory effect on MMP is associated with reduced mechanical allodynia. CONCLUSIONS: This study identifies NOV as a new actor against inflammatory pain through regulation of MMPs thus uncovering NOV as an attractive candidate for therapeutic improvement in pain relief.


Immediate-Early Proteins/metabolism , Inflammation/complications , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Pain/etiology , Pain/metabolism , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Chemokine CCL2/metabolism , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Freund's Adjuvant , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Immediate-Early Proteins/genetics , Inflammation/chemically induced , Intercellular Signaling Peptides and Proteins/genetics , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Nerve Tissue Proteins/metabolism , Pain/drug therapy , Pain Measurement , Pain Threshold/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Spinal Cord/pathology , Time Factors , Transfection , Up-Regulation/drug effects
5.
Dev Neurobiol ; 72(11): 1363-75, 2012 Nov.
Article En | MEDLINE | ID: mdl-22038708

NOV/CCN3 is one of the founding members of the CCN (Cyr61 CTGF NOV) family. In the avian retina, CCN3 expression is mostly located within the central region of the inner nuclear layer. As retinal development progresses and this retinal layer differentiates and matures, CCN3 expression forms a dorsal-ventral and a central-peripheral gradient. CCN3 is produced by two glial cell types, peripapillary cells and Müller cells, as well as by horizontal, amacrine, and bipolar interneurons. In retinal neurons and Müller cell cultures, CCN3 expression is induced by activated BMP signaling, whereas Notch signaling decreases CCN3 mRNA and protein levels in Müller cells and has no effect in retinal neurons. In Müller cells, the CCN3 expression detected may thus result from a balance between the Notch and BMP signaling pathways.


Nephroblastoma Overexpressed Protein/metabolism , RNA, Messenger/analysis , Retina , Retinal Neurons/metabolism , Animals , Bone Morphogenetic Protein Receptors/metabolism , Cells, Cultured , Chick Embryo , Gene Expression Regulation, Developmental/physiology , Immunohistochemistry , Neuroglia/metabolism , Receptor, Notch1/metabolism , Retina/embryology , Retina/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
6.
J Clin Endocrinol Metab ; 92(8): 3253-60, 2007 Aug.
Article En | MEDLINE | ID: mdl-17566092

CONTEXT: Childhood adrenocortical tumors (ACTs) have a fetal adrenal phenotype and overexpress steroidogenic factor-1 (SF-1). Nephroblastoma overexpressed (NOV)/cysteine-rich protein 61/connective tissue growth factor/nephroblastoma overexpressed gene-3 mRNA is significantly down-regulated in childhood ACTs. OBJECTIVE: The objective of the study was to measure NOV protein levels in childhood ACTs and characterize NOV expression regulation and biological function in human adrenocortical cells. DESIGN AND SETTING: Protein extracts from ACT and normal adrenal cortex samples, human adrenocortical carcinoma H295R, primary adrenocortical tumors and fetal adrenal cultures, tissue culture supernatants, and cell lysates from H295R cells overexpressing SF-1 in an inducible fashion were used. MAIN OUTCOME MEASURES: NOV protein levels were measured by enzyme-linked immunoassay and immunoblot. Transient transfection assays were used to study the activity of NOV promoter. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling, caspase assays, and flow cytometry were used to assess the proapoptotic activity of NOV on cells in culture. RESULTS: NOV mRNA and protein expression is lower in childhood ACTs than in normal adrenal cortex. No significant difference was observed between adenomas and carcinomas. SF-1 overexpression down-regulates NOV at the transcriptional level. NOV has a selective proapoptotic activity toward human adrenocortical cells. The C-terminal domain of NOV is responsible for its proapoptotic effect. NOV protein is expressed in DAX-1-positive human fetal adrenal cells. CONCLUSIONS: NOV is a selective proapoptotic factor for human adrenocortical cells. Reduced expression of NOV in ACTs may play an important role in the process of childhood ACT tumorigenesis, accounting at least in part for the defect of apoptotic regression of the fetal adrenal that has been proposed to be responsible for tumor formation.


Adenoma/metabolism , Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex/cytology , Apoptosis/genetics , Apoptosis/physiology , Carcinoma/metabolism , Gene Expression Regulation, Neoplastic/genetics , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Adenoma/genetics , Adenoma/pathology , Adrenal Cortex/physiology , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/pathology , Carcinoma/genetics , Carcinoma/pathology , Caspases/metabolism , Cell Line, Tumor , Child , Connective Tissue Growth Factor , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Down-Regulation/genetics , Down-Regulation/physiology , Enzyme Activation/physiology , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/physiology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/biosynthesis , Immunoblotting , Intercellular Signaling Peptides and Proteins/biosynthesis , Luciferases/biosynthesis , Luciferases/genetics , Nephroblastoma Overexpressed Protein , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroidogenic Factor 1 , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
7.
Lab Invest ; 87(7): 680-9, 2007 Jul.
Article En | MEDLINE | ID: mdl-17496904

Remodeling of extracellular matrix (ECM) is an important physiological feature of normal growth and development. Recent studies have emphasized the role of matrix metalloproteinases (MMP-2 and MMP-9) in normal mouse nephrogenesis. We have demonstrated previously in the rat that in utero exposure to maternal diabetes impairs renal development leading to a 30% reduction in the nephron number. Transforming growth factor-beta1 (TGF-beta1) and connective tissue growth factor (CTGF) are known to mediate high glucose effects on matrix degradation. The aim of the present study was to address the expression of type IV collagenase and TGF-beta1/CTGF systems in rat kidney during normal development and after in utero exposure to maternal diabetes. Both MMP-2 and MMP-9 mRNA metanephric expressions and activities were dramatically downregulated in kidneys issued from diabetic fetuses and in metanephros cultured in the presence of high glucose concentration. TGF-beta1 and CTGF expressions were significantly enhanced in diabetic fetal kidneys and in high glucose cultured metanephroi. Conditioned media obtained from metanephroi grown with high glucose concentration upregulated functional TGF-beta activity in transfected ATDC5 cells. In conclusion, in impaired nephrogenesis resulting from in utero exposure to maternal diabetes, alteration of both type IV collagenase and TGF-beta1/CTGF systems may lead to abnormal remodeling of ECM, which may, in turn, induce defects in ureteral bud branching leading to the observed reduction in the nephron number with consequences later in life: progression of chronic renal disease and hypertension.


Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Kidney/embryology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Organogenesis/physiology , Pregnancy in Diabetics/metabolism , Animals , Cells, Cultured , Connective Tissue Growth Factor , Diabetes Mellitus, Experimental/chemically induced , Extracellular Matrix/chemistry , Extracellular Matrix/enzymology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , In Situ Hybridization , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Staining and Labeling , Transforming Growth Factor beta1/metabolism
8.
Exp Cell Res ; 312(10): 1876-89, 2006 Jun 10.
Article En | MEDLINE | ID: mdl-16600215

NOV (nephroblastoma overexpressed) is a member of a family of proteins which encodes secreted matrix-associated proteins. NOV is expressed during development in dermomyotome and limb buds, but its functions are still poorly defined. In order to understand the role of NOV in myogenic differentiation, C2C12 cells overexpressing NOV (C2-NOV) were generated. These cells failed to engage into myogenic differentiation, whereas they retained the ability to differentiate into osteoblasts. In differentiating conditions, C2-NOV cells remained proliferative, failed to express differentiation markers and lost their ability to form myotubes. Inhibition of differentiation by NOV was also observed with human primary muscle cells. Further examination of C2-NOV cells revealed a strong downregulation of the myogenic determination genes MyoD and Myf5 and of IGF-II expression. MyoD forced expression in C2-NOV was sufficient to restore differentiation and IGF-II induction whereas 10(-6) M insulin treatment had no effects. NOV therefore acts upstream of MyoD and does not affect IGF-II induction and signaling. HES1, a target of Notch, previously proposed to mediate NOV action, was not implicated in the inhibition of differentiation. We propose that NOV is a specific cell fate regulator in the myogenic lineage, acting negatively on key myogenic genes thus controlling the transition from progenitor cells to myoblasts.


Cell Differentiation/physiology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle Cells/physiology , Muscle, Skeletal/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cells, Cultured , Connective Tissue Growth Factor , Culture Media/chemistry , Genes, Reporter , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immediate-Early Proteins/genetics , Insulin/metabolism , Insulin-Like Growth Factor II/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mice , Morphogenesis , Muscle Cells/cytology , Muscle, Skeletal/cytology , MyoD Protein/genetics , MyoD Protein/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Nephroblastoma Overexpressed Protein , Receptors, Notch/metabolism , Signal Transduction/physiology , Transcription Factor HES-1
9.
Cell Commun Adhes ; 12(1-2): 41-57, 2005.
Article En | MEDLINE | ID: mdl-16371345

During mammalian development, expression of the Nephroblastoma overexpressed gene (NOV/CCN3) is tightly regulated in skeletal muscles. Ex vivo, ectopic expression of NOV blocks myogenic differentiation. NOV also supports endothelial cell adhesion and angiogenesis through interactions with integrins. Integrins play fundamental roles during myogenesis. In this study, we show that NOV mediates adhesion and spreading of myoblasts. Myoblasts adhesion to NOV does not require proteoglycans and is dependent on integrin beta1, whereas spreading involves another RGD-sensitive integrin. The C-Terminal part of NOV as well as full-length is able to support adhesion of myoblasts; in addition, both increase focal-adhesion kinase (FAK) phosphorylation. Furthermore, NOV is an adhesive substrate that, combined with FGF2 or IGF-1, promotes cell specific proliferation and survival, respectively, in a better way than fibronectin. Taken together, these results identify NOV as an adhesion substrate for myoblasts which, in concert with growth factors, could play a role in the physiology of muscle cells.


Fibroblast Growth Factor 2/pharmacology , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Animals , Apoptosis , Cell Adhesion , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Connective Tissue Growth Factor , Cytoskeleton/metabolism , DNA/biosynthesis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Immediate-Early Proteins/chemistry , Insulin-Like Growth Factor I/pharmacology , Integrins/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Kinetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Myoblasts/cytology , Nephroblastoma Overexpressed Protein , Phosphorylation , Poly(ADP-ribose) Polymerases/metabolism , Proteoglycans/metabolism
10.
J Bone Miner Res ; 20(12): 2213-23, 2005 Dec.
Article En | MEDLINE | ID: mdl-16294274

UNLABELLED: We studied the involvement of NOV/CCN3, whose function is poorly understood, in chondrocyte differentiation. NOV was found to upregulate TGF-beta2 and type X collagen and to act as a downstream effector of TGF-beta1 in ATDC5 and primary chondrocytes. Thus, NOV is a positive modulator of chondrogenesis. INTRODUCTION: NOV/CCN3 is a matricellular protein that belongs to the CCN family. A growing body of evidence indicates that NOV could play a role in cell differentiation, particularly in chondrogenesis. During chick embryo development, NOV expression is tightly regulated in cartilage, and a high expression of NOV has been associated with cartilage differentiation in Wilms' tumors. However, a precise role for NOV and potential target genes of NOV in chondrogenesis are unknown. MATERIALS AND METHODS: ATDC5 cells and primary chondrocytes were either treated with NOV recombinant protein or transfected with a NOV-specific siRNA to determine, using quantitative RT-PCR, the effect of NOV on the expression of several molecules involved in chondrocyte differentiation. Stable ATDC5 clones expressing NOV were also established to show that NOV was a downstream effector of TGF-beta1. RESULTS: We established that NOV/CCN3 expression increases in ATDC5 cells at early stages of chondrogenic differentiation and precedes the appearance of TGF-beta2 and of several chondrocytic markers such as SOX9 or type X collagen. When exogenously administered, NOV recombinant protein up-regulates TGF-beta2 and type X collagen mRNA levels both in ATDC5 cells and in primary mouse chondrocytes but does not influence SOX9 expression. This regulation also occurs at the endogenous level because downregulation of NOV expression is correlated with an inhibition of TGF-beta2 and type X collagen in primary chondrocytes. Furthermore, we found that NOV expression is downregulated when chondrocytes are exposed to TGF-beta1-dedifferentiating treatment in chondrocytes, further providing evidence that NOV may counteract TGF-beta1 effects on chondrocytes. CONCLUSIONS: This study provides the first characterization of two new targets of NOV involved in chondrocyte differentiation, shows that NOV acts with TGF-beta1 in a cascade of gene regulation, and indicates that NOV is a positive modulator of chondrogenesis.


Chondrocytes/metabolism , Collagen Type X/genetics , Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Transforming Growth Factor beta/genetics , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Collagen Type II/genetics , Connective Tissue Growth Factor , Gene Expression/drug effects , High Mobility Group Proteins/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/pharmacology , Insulin/pharmacology , Integrin alpha5beta1/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Nephroblastoma Overexpressed Protein , Proteins/pharmacology , RNA, Small Interfering/genetics , SOX9 Transcription Factor , Transcription Factors/genetics , Transfection , Transforming Growth Factor beta2
11.
J Biol Chem ; 277(43): 41220-9, 2002 Oct 25.
Article En | MEDLINE | ID: mdl-12149257

The human NOV secreted glycoprotein (NOVH) is abundant in the fetal and adult adrenal cortex. The amount of NOVH increases in benign adrenocortical tumors and decreases in malignant adrenocortical tumors, suggesting that NOVH plays a role in tumorigenesis in the adrenal cortex. Transforming growth factor beta1 (TGFbeta1), fibroblast growth factor 2 (FGF2), and insulin growth factors (IGFs) play crucial roles in the physiology of the adrenal cortex. We investigated the effects of these factors on the expression of novH in the NCI H295R adrenocortical cell line. The amounts of NOVH protein and novH transcripts were down-regulated by TGFbeta1 and up-regulated by FGF2, whereas IGFs had no effect. Furthermore, the TGFbeta1-dependent inhibition of novH promoter activity was completely abrogated following site-directed mutation of two activating protein (AP-1) sequences (positions -473 and -447), whereas the stimulatory effect of FGF2 was not affected. Co-transfection with dominant negative forms of c-Jun and MEKK1 also abrogated novH-targeted regulation by TGFbeta1, whereas the overproduction of Smad proteins or dominant negative forms of Smad had no effect. Taken together, these results suggest that c-Jun and MEKK1 signaling but not Smad signaling are involved in the TGFbeta1-dependent decrease in NOVH in NCI H295R cells. In conclusion, our data provide evidence that novH is a new target of TGFbeta1; unlike other members of the CCN (cyr61, ctgf, nov) family, however, its expression is repressed rather than induced.


Adrenal Cortex/metabolism , Down-Regulation/physiology , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins c-jun/physiology , Transforming Growth Factor beta/physiology , Adrenal Cortex/cytology , Base Sequence , Cell Line , Connective Tissue Growth Factor , DNA , Fibroblast Growth Factor 2/physiology , Molecular Sequence Data , Nephroblastoma Overexpressed Protein , Promoter Regions, Genetic , Sequence Homology, Nucleic Acid , Up-Regulation
...