1
|
Fadhil Jaafar A, Afrisham R, Fadaei R, Farrokhi V, Moradi N, Abbasi A, Einollahi N. CCN3/NOV serum levels in coronary artery disease (CAD) patients and its correlation with TNF-α and IL-6. BMC Res Notes 2023; 16:306. [PMID: 37919772 PMCID: PMC10623743 DOI: 10.1186/s13104-023-06590-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023] Open
Abstract
INTRODUCTION Dysregulation in the secretion of adipokines or adipocytokines plays a significant role in triggering a pro-inflammatory state, leading to endothelial dysfunction and insulin resistance, and ultimately elevating the risk of atherosclerosis and coronary artery disease (CAD). Previous studies have shown a link between NOV/CCN3 (an adipokine) and obesity, insulin resistance, and inflammation. However, no research has explored the relationship between CCN3 serum levels and CAD. Therefore, we conducted the first investigation to examine the correlation between CCN3 and CAD risk factors in patients. METHODS In a case-control study, we measured the serum levels of CCN3, IL-6, adiponectin, and TNF-α in 88 angiography-confirmed CAD patients and 88 control individuals using ELISA kits. Additionally, we used an auto analyzer and commercial kits to measure the biochemical parameters. RESULTS In patients with CAD, the serum levels of CCN3, TNF-α, and IL-6 were significantly higher compared to the control group, whereas lower levels of adiponectin were observed in the CAD group (P < 0.0001). A positive correlation was found between CCN3 and IL-6 and TNF-α in the CAD group ([r = 0.38, P < 0.0001], [r = 0.39, P < 0.0001], respectively). A binary logistic regression analysis showed the risk of CAD in the model adjusted (OR [95% CI] = 1.29 [1.19 - 1.41]), (P < 0.0001). We determined a cut-off value of CCN3 (3169.6 pg/mL) to distinguish CAD patients from the control group, with good sensitivity and specificity obtained for this finding (83.8% and 87.5%, respectively). CONCLUSION This study provides evidence of a positive association between CCN3 serum levels and CAD, as well as inflammation markers such as IL-6 and TNF-α. These findings suggest that CCN3 may serve as a potential biomarker for CAD, and further investigations are necessary to validate this association and explore its potential use in clinical settings.
Collapse
Affiliation(s)
- Alaa Fadhil Jaafar
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Afrisham
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Fadaei
- Sleep Disorders Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Vida Farrokhi
- Department of Hematology, Faculty of Allied Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nariman Moradi
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Abbasi
- Department of Cardiology, Dr Shariatee training and research Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Nahid Einollahi
- Department of Clinical Laboratory Sciences, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Dittmann KH, Mayer C, Stephan H, Mieth C, Bonin M, Lechmann B, Rodemann HP. Exposure of primary osteoblasts to combined magnetic and electric fields induced spatiotemporal endochondral ossification characteristic gene- and protein expression profiles. J Exp Orthop 2022; 9:39. [PMID: 35499653 PMCID: PMC9061914 DOI: 10.1186/s40634-022-00477-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022] Open
Abstract
Purpose Molecular processes in primary osteoblasts were analyzed in response to magnetic and electric field exposure to examine its potential impact on bone healing. Methods Primary osteoblasts were exposed to a combination of a magnetic field and an additional electric field (EFMF) (20 Hz, 700 mV, 5 mT, continuous sinusoids) in vitro. mRNA- and protein-expressions were assessed during a time interval of 21 days and compared with expression data obtained from control osteoblasts. Results We observed an autonomous osteoblast differentiation process in vitro under the chosen cultivation conditions. The initial proliferative phase was characterized by a constitutively high mRNA expression of extracellular matrix proteins. Concurrent EFMF exposure resulted in significanly increased cell proliferation (fold change: 1.25) and reduced mRNA-expressions of matrix components (0.5–0.75). The following reorganization of the extracellular matrix is prerequisite for matrix mineralization and is characterised by increased Ca2+ deposition (1.44). On molecular level EFMF exposure led to a significant decreased thrombospondin 1 (THBS1) mRNA- (0.81) and protein- (0.54) expression, which in turn reduced the TGFß1-dependent mRNA- (0.68) and protein- (0.5) expression of transforming growth factor beta induced (ßIG-H3) significantly, an inhibitor of endochondral ossification. Consequently, EFMF exposure stimulated the expression of genes characteristic for endochondral ossification, such as collagen type 10, A1 (1.50), osteopontin (1.50) and acellular communication network factor 3 (NOV) (1.45). Conclusions In vitro exposure of osteoblasts to EFMF supports cell differentiation and induces gene- and protein-expression patterns characteristic for endochondral ossification during bone fracture healing in vivo. Supplementary Information The online version contains supplementary material available at 10.1186/s40634-022-00477-9.
Collapse
|
3
|
CCN proteins in the musculoskeletal system: current understanding and challenges in physiology and pathology. J Cell Commun Signal 2021; 15:545-566. [PMID: 34228239 PMCID: PMC8642527 DOI: 10.1007/s12079-021-00631-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The acronym for the CCN family was recently revised to represent “cellular communication network”. These six, small, cysteine-enriched and evolutionarily conserved proteins are secreted matricellular proteins, that convey and modulate intercellular communication by interacting with structural proteins, signalling factors and cell surface receptors. Their role in the development and physiology of musculoskeletal system, constituted by connective tissues where cells are interspersed in the cellular matrix, has been broadly studied. Previous research has highlighted a crucial balance of CCN proteins in mesenchymal stem cell commitment and a pivotal role for CCN1, CCN2 and their alter ego CCN3 in chondrogenesis and osteogenesis; CCN4 plays a minor role and the role of CCN5 and CCN6 is still unclear. CCN proteins also participate in osteoclastogenesis and myogenesis. In adult life, CCN proteins serve as mechanosensory proteins in the musculoskeletal system providing a steady response to environmental stimuli and participating in fracture healing. Substantial evidence also supports the involvement of CCN proteins in inflammatory pathologies, such as osteoarthritis and rheumatoid arthritis, as well as in cancers affecting the musculoskeletal system and bone metastasis. These matricellular proteins indeed show involvement in inflammation and cancer, thus representing intriguing therapeutic targets. This review discusses the current understanding of CCN proteins in the musculoskeletal system as well as the controversies and challenges associated with their multiple and complex roles, and it aims to link the dispersed knowledge in an effort to stimulate and guide readers to an area that the writers consider to have significant impact and relevant potentialities.
Collapse
|
4
|
Henrot P, Moisan F, Laurent P, Manicki P, Kaulanjan-Checkmodine P, Jolivel V, Rezvani HR, Leroy V, Picard F, Boulon C, Schaeverbeke T, Seneschal J, Lazaro E, Taïeb A, Truchetet ME, Cario M. Decreased CCN3 in Systemic Sclerosis Endothelial Cells Contributes to Impaired Angiogenesis. J Invest Dermatol 2020; 140:1427-1434.e5. [PMID: 31954725 DOI: 10.1016/j.jid.2019.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 11/30/2022]
Abstract
Systemic sclerosis (SSc) is a rare and severe connective tissue disease combining autoimmune and vasculopathy features, ultimately leading to organ fibrosis. Impaired angiogenesis is an often silent and life-threatening complication of the disease. We hypothesize that CCN3, a member of the CCN family of extracellular matrix proteins, which is an antagonist of the profibrotic protein CCN2 as well as a proangiogenic factor, is implicated in SSc pathophysiology. We performed skin biopsies on 26 patients with SSc, both in fibrotic and nonfibrotic areas for 17 patients, and collected 18 healthy control skin specimens for immunohistochemistry and cell culture. Histological analysis of nonfibrotic and fibrotic SSc skin shows a systemic decrease of papillary dermis surface as well as disappearance of capillaries. CCN3 expression is systematically decreased in the dermis of patients with SSc compared with healthy controls, particularly in dermal blood vessels. Moreover, CCN3 is decreased in vitro in endothelial cells from patients with SSc. We show that CCN3 is essential for endothelial cell migration and angiogenesis in vitro. In conclusion, CCN3 may represent a promising therapeutic target for patients with SSc presenting with vascular involvement.
Collapse
Affiliation(s)
- Pauline Henrot
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France; Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France.
| | - François Moisan
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France
| | - Paôline Laurent
- University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Pauline Manicki
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France; University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | | | - Valérie Jolivel
- University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Hamid Reza Rezvani
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France; Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| | - Vaianu Leroy
- Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| | - François Picard
- Department of Cardiology, Hôpital Haut-Levêque, Pessac, France
| | - Carine Boulon
- Department of Vascular Medicine, Hôpital Saint André, Bordeaux, France
| | - Thierry Schaeverbeke
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France
| | - Julien Seneschal
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France; Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| | - Estibaliz Lazaro
- University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France; Department of Internal Medicine, National Reference Center for Systemic Autoimmune Rare Diseases, Hôpital Haut-Levêque, Pessac, France
| | - Alain Taïeb
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France; Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| | - Marie-Elise Truchetet
- Department of Rheumatology, National Reference Center for Systemic Autoimmune Rare Diseases, Hopital Pellegrin, Bordeaux, France; University of Bordeaux, CNRS, Immunoconcept, UMR 5164, Bordeaux, France
| | - Muriel Cario
- University of Bordeaux, Inserm, BMGIC, UMR1035, Bordeaux, France; Department of Dermatology and Pediatric Dermatology, National Center for Rare Skin Disorders, Hôpital Saint André, Bordeaux, France
| |
Collapse
|
5
|
de la Vega Gallardo N, Dittmer M, Dombrowski Y, Fitzgerald DC. Regenerating CNS myelin: Emerging roles of regulatory T cells and CCN proteins. Neurochem Int 2018; 130:104349. [PMID: 30513363 DOI: 10.1016/j.neuint.2018.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
Abstract
Efficient myelin regeneration in the central nervous system (CNS) requires the migration, proliferation and differentiation of oligodendrocyte progenitor cells (OPC) into myelinating oligodendrocytes. In demyelinating diseases such as multiple sclerosis (MS), this regenerative process can fail, and therapies targeting myelin repair are currently completely lacking in the clinic. The immune system is emerging as a key regenerative player in many tissues, such as muscle and heart. We recently reported that regulatory T cells (Treg) are required for efficient CNS remyelination. Furthermore, Treg secrete CCN3, a matricellular protein from the CCN family, implicated in regeneration of other tissues. Treg-derived CCN3 promoted oligodendrocyte differentiation and myelination. In contrast, previous studies showed that CCN2 inhibited myelination. These studies highlight the need for further scrutiny of the roles that CCN proteins play in myelin development and regeneration. Collectively, these findings open up exciting avenues of research to uncover the regenerative potential of the adaptive immune system.
Collapse
Affiliation(s)
- Nira de la Vega Gallardo
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Marie Dittmer
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Northern Ireland, UK.
| |
Collapse
|
6
|
Kong Y, Guo Y, Zhang J, Zhao B, Wang J. Strontium Promotes Transforming Growth Factors β1 and β2 Expression in Rat Chondrocytes Cultured In Vitro. Biol Trace Elem Res 2018; 184:450-455. [PMID: 29170863 DOI: 10.1007/s12011-017-1208-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 01/18/2023]
Abstract
The transforming growth factors β1 (TGF-β1) and TGF-β2, as two distinct homodimers of TGF-β superfamily, involve in chondrocyte growth and differentiation. Emerging evidence has implied that strontium (Sr) plays an important role in the bone formation and resorption, and has strong effects on stimulating human cartilage matrix formation in vitro. However, the direct effects of Sr on TGF-β1 and TGF-β2 expressions in chondrocytes are not entirely clear. The purpose of this study was to evaluate the influence of different Sr concentrations on the expression of TGF-β1 and TGF-β2 in rat chondrocytes in vitro. Chondrocytes were isolated from Wistar rat articular by enzymatic digestion. Strontium chloride hexahydrate (SrCl2·6H2O) was used as a Sr source in this study. Sr was added to the culture solution at final concentrations of 0, 0.5, 1.0, 2.0, 5.0, 20.0, and 100 μg/mL. After 72 h of continuous culture, TGF-β1 and TGF-β2 mRNA abundance and protein expression levels in the chondrocytes were determined by real-time polymerase chain reaction (real-time PCR) and Western blot, respectively. The results showed that TGF-β1 and TGF-β2 expressions in chondrocytes increased dose-dependently with Sr concentration. The mRNA abundance of TGF-β1 and TGF-β2 were markedly higher than those observed for control (P < 0.01) when the Sr-treated concentration exceeded 1.0 and 5.0 μg/mL, respectively. The TGF-β1 and TGF-β2 protein expression levels were extremely significantly higher than those in the control group (P < 0.01) at above 5.0 μg/mL Sr-treatment. These results indicated that Sr could involve in the chondrocytes metabolism via regulating TGF-β1 and TGF-β2 signalling.
Collapse
Affiliation(s)
- Yezi Kong
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yazhou Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jinfeng Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
7
|
Martinerie C, Garcia M, Do TTH, Antoine B, Moldes M, Dorothee G, Kazazian C, Auclair M, Buyse M, Ledent T, Marchal PO, Fesatidou M, Beisseiche A, Koseki H, Hiraoka S, Chadjichristos CE, Blondeau B, Denis RG, Luquet S, Fève B. NOV/CCN3: A New Adipocytokine Involved in Obesity-Associated Insulin Resistance. Diabetes 2016; 65:2502-15. [PMID: 27284105 DOI: 10.2337/db15-0617] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 05/23/2016] [Indexed: 11/13/2022]
Abstract
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.
Collapse
Affiliation(s)
- Cécile Martinerie
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Marie Garcia
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Thi Thu Huong Do
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Bénédicte Antoine
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Marthe Moldes
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Guillaume Dorothee
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France
| | - Chantal Kazazian
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Martine Auclair
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Marion Buyse
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France Department of Pharmacy, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France Paris-Sud University, EA 4123, Châtenay-Malabry, France
| | - Tatiana Ledent
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France
| | - Pierre-Olivier Marchal
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Maria Fesatidou
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Adrien Beisseiche
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Cordeliers Research Center, Paris, France
| | - Haruhiko Koseki
- RIKEN Research Center for Allergy and Immunology (RCAI), RIKEN Yokohama Institute, Yokohama, Japan
| | - Shuichi Hiraoka
- Department of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | | | - Bertrand Blondeau
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Cordeliers Research Center, Paris, France
| | | | - Serge Luquet
- Sorbonne Paris City University, Paris Diderot University, BFA, CNRS, Paris, France
| | - Bruno Fève
- Sorbonne Universities, Pierre and Marie Curie University Paris 06, INSERM, Saint-Antoine Research Center, Saint-Antoine Hospital, Paris, France Hospitalo-Universitary Institute, ICAN, Paris, France Department of Endocrinology, Paris, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, Paris, France
| |
Collapse
|
8
|
Yoshioka Y, Ono M, Maeda A, Kilts TM, Hara ES, Khattab H, Ueda J, Aoyama E, Oohashi T, Takigawa M, Young MF, Kuboki T. CCN4/WISP-1 positively regulates chondrogenesis by controlling TGF-β3 function. Bone 2016; 83:162-170. [PMID: 26555637 PMCID: PMC5749225 DOI: 10.1016/j.bone.2015.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/06/2015] [Accepted: 11/06/2015] [Indexed: 01/05/2023]
Abstract
The CCN family of proteins plays important roles in development and homeostasis of bone and cartilage. To understand the role of CCN4 in chondrogenesis, human bone marrow stromal cells (hBMSCs) were transduced with CCN4 adenovirus (adCCN4) or siRNA to CCN4 (siCCN4) in the presence or absence of transforming growth factor-β3 (TGF-β3). Overexpression of CCN4 enhanced TGF-β3-induced SMAD2/3 phosphorylation and chondrogenesis of hBMSCs in an in vitro assay using a micromass culture model. On the other hand, knockdown of CCN4 inhibited the TGF-β3-induced SMAD2/3 phosphorylation and synthesis of cartilage matrix in micromass cultures of hBMSCs. Immunoprecipitation-western blot analysis revealed that CCN4 bound to TGF-β3 and regulated the ability of TGF-β3 to bind to hBMSCs. In vivo analysis confirmed there was a significant decrease in the gene expression levels of chondrocyte markers in cartilage samples from Ccn4-knock out (KO) mice, compared to those from wild type (WT) control. In order to investigate the regenerative properties of the articular cartilage in Ccn4-KO mice, articular cartilage defects were surgically performed in the knee joints of young mice, and the results showed that the cartilage was partially repaired in WT mice, but not in Ccn4-KO mice. In conclusion, these results show, for the first time, that CCN4 has a positive influence on chondrogenic differentiation by modulating the effects of TGF-β3.
Collapse
Affiliation(s)
- Yuya Yoshioka
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuaki Ono
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Azusa Maeda
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Tina M Kilts
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Emilio Satoshi Hara
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hany Khattab
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan; Advanced Research Center for Oral & Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Junji Ueda
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Eriko Aoyama
- Advanced Research Center for Oral & Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshitaka Oohashi
- Advanced Research Center for Oral & Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masaharu Takigawa
- Advanced Research Center for Oral & Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
9
|
RNA Seq profiling reveals a novel expression pattern of TGF-β target genes in human blood eosinophils. Immunol Lett 2015; 167:1-10. [PMID: 26112417 DOI: 10.1016/j.imlet.2015.06.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 12/16/2022]
Abstract
Despite major advances in our understanding of TGF-β signaling in multiple cell types, little is known about the direct target genes of this pathway in human eosinophils. These cells constitute the major inflammatory component present in the sputum and lung of active asthmatics and their numbers correlate well with disease severity. During the transition from acute to chronic asthma, TGF-β levels rise several fold in the lung which drives fibroblasts to produce extracellular matrix (ECM) and participate in airway and parenchymal remodeling. In this report, we use purified blood eosinophils from healthy donors and analyze baseline and TGF-β responsive genes by RNA Seq, and demonstrate that eosinophils (PBE) express 7981 protein-coding genes of which 178 genes are up-regulated and 199 genes are down-regulated by TGF-β. While 18 target genes have been previously associated with asthma and eosinophilic disorders, the vast majority have been implicated in cell death and survival, differentiation, and cellular function. Ingenuity pathway analysis revealed that 126 canonical pathways are activated by TGF-β including iNOS, TREM1, p53, IL-8 and IL-10 signaling. As TGF-β is an important cytokine for eosinophil function and survival, and pulmonary inflammation and fibrosis, our results represent a significant step toward the identification of novel TGF-β responsive genes and provide a potential therapeutic opportunity by selectively targeting relevant genes and pathways.
Collapse
|
10
|
Zhang P, Ravuri SK, Wang J, Marra KG, Kling RE, Chai J. Exogenous connective tissue growth factor preserves the hair-inductive ability of human dermal papilla cells. Int J Cosmet Sci 2014; 36:442-50. [PMID: 24925376 DOI: 10.1111/ics.12146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 05/28/2014] [Indexed: 01/14/2023]
Abstract
Connective tissue growth factor influences human dermal papilla cells' hair inductive ability through several signaling pathways.
Collapse
Affiliation(s)
- P Zhang
- Medical School of Chinese People's Liberation Army, #28 Fuxing Road, Haidian District, Beijing, 100853, China; Department of Burns and Plastic Surgery, First Hospital Affiliated to General Hospital of PLA, #51 Fucheng Road, Haidian District, Beijing, 100048, China
| | | | | | | | | | | |
Collapse
|
11
|
Kubota S, Takigawa M. The CCN family acting throughout the body: recent research developments. Biomol Concepts 2013; 4:477-94. [DOI: 10.1515/bmc-2013-0018] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 07/24/2013] [Indexed: 12/26/2022] Open
Abstract
AbstractThe animal body is composed of a variety of cells and extracellular matrices that are organized and orchestrated in a harmonized manner to support life. Therefore, the critical importance of a comprehensive understanding of the molecular network surrounding and integrating the cells is now emphasized. The CCN family is a novel group of matricellular proteins that interact with and orchestrate a number of extracellular signaling and matrix molecules to construct and maintain living tissues. This family comprises six distinct members in mammals, which are characterized by a unique and conserved modular structure. These proteins are not targeted to limited and specific receptors to execute specific missions, but manipulate a vast number of biomolecules in the network by serving as a molecular hub at the center. The unified nomenclature, CCN, originates from a simple acronym of the three classical members, which helps us to avoid having any preconception about their pleiotropic and anonymous functional nature. In this review, after a brief summary of the general molecular concepts regarding the CCN family, new aspects of each member uncovered by recent research are introduced, which represent, nevertheless, only the tip of the iceberg of the profound functionality of these molecules.
Collapse
Affiliation(s)
- Satoshi Kubota
- 1Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, and Okayama University Dental School Advanced Research Center for Oral and Craniofacial Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| | - Masaharu Takigawa
- 1Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, and Okayama University Dental School Advanced Research Center for Oral and Craniofacial Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8525, Japan
| |
Collapse
|
12
|
Matsushita Y, Sakamoto K, Tamamura Y, Shibata Y, Minamizato T, Kihara T, Ito M, Katsube KI, Hiraoka S, Koseki H, Harada K, Yamaguchi A. CCN3 protein participates in bone regeneration as an inhibitory factor. J Biol Chem 2013; 288:19973-85. [PMID: 23653360 DOI: 10.1074/jbc.m113.454652] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
CCN3, a member of the CCN protein family, inhibits osteoblast differentiation in vitro. However, the role of CCN3 in bone regeneration has not been well elucidated. In this study, we investigated the role of CCN3 in bone regeneration. We identified the Ccn3 gene by microarray analysis as a highly expressed gene at the early phase of bone regeneration in a mouse bone regeneration model. We confirmed the up-regulation of Ccn3 at the early phase of bone regeneration by RT-PCR, Western blot, and immunofluorescence analyses. Ccn3 transgenic mice, in which Ccn3 expression was driven by 2.3-kb Col1a1 promoter, showed osteopenia compared with wild-type mice, but Ccn3 knock-out mice showed no skeletal changes compared with wild-type mice. We analyzed the bone regeneration process in Ccn3 transgenic mice and Ccn3 knock-out mice by microcomputed tomography and histological analyses. Bone regeneration in Ccn3 knock-out mice was accelerated compared with that in wild-type mice. The mRNA expression levels of osteoblast-related genes (Runx2, Sp7, Col1a1, Alpl, and Bglap) in Ccn3 knock-out mice were up-regulated earlier than those in wild-type mice, as demonstrated by RT-PCR. Bone regeneration in Ccn3 transgenic mice showed no significant changes compared with that in wild-type mice. Phosphorylation of Smad1/5 was highly up-regulated at bone regeneration sites in Ccn3 KO mice compared with wild-type mice. These results indicate that CCN3 is up-regulated in the early phase of bone regeneration and acts as a negative regulator for bone regeneration. This study may contribute to the development of new strategies for bone regeneration therapy.
Collapse
Affiliation(s)
- Yuki Matsushita
- Section of Oral Pathology, International Research Center for Molecular Science in Tooth and Bone Diseases, Tokyo Medical and Dental University, Tokyo 113-8549, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Tran CM, Smith HE, Symes A, Rittié L, Perbal B, Shapiro IM, Risbud MV. Transforming growth factor β controls CCN3 expression in nucleus pulposus cells of the intervertebral disc. ACTA ACUST UNITED AC 2013; 63:3022-31. [PMID: 21618206 DOI: 10.1002/art.30468] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE To investigate transforming growth factor β (TGFβ) regulation of CCN3 expression in cells of the nucleus pulposus. METHODS Real-time reverse transcription-polymerase chain reaction and Western blot analyses were used to measure CCN3 expression in the nucleus pulposus. Transfections were used to measure the effect of Smad3, MAPKs, and activator protein 1 (AP-1) on TGFβ-mediated CCN3 promoter activity. Lentiviral knockdown of Smad3 was performed to assess the role of Smad3 in CCN3 expression. RESULTS CCN3 was expressed in embryonic and adult intervertebral discs. TGFβ decreased the expression of CCN3 and suppressed its promoter activity in nucleus pulposus cells. DN-Smad3, Smad3 small interfering RNA, or DN-AP-1 had little effect on TGFβ suppression of CCN3 promoter activity. However, p38 and ERK inhibitors blocked suppression of CCN3 by TGFβ, suggesting involvement of these signaling pathways in the regulation of CCN3. Interestingly, overexpression of Smad3 in the absence of TGFβ increased CCN3 promoter activity. We validated the role of Smad3 in controlling CCN3 expression in Smad3-null mice and in nucleus pulposus cells transduced with lentiviral short hairpin Smad3. In terms of function, treatment with recombinant CCN3 showed a dose-dependent decrease in the proliferation of nucleus pulposus cells. Moreover, CCN3-treated cells showed a decrease in aggrecan, versican, CCN2, and type I collagen expression. CONCLUSION The opposing effect of TGFβ on CCN2 and CCN3 expression and the suppression of CCN2 by CCN3 in nucleus pulposus cells further the paradigm that these CCN proteins form an interacting triad, which is possibly important in maintaining extracellular matrix homeostasis and cell numbers.
Collapse
Affiliation(s)
- Cassie M Tran
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Laurent M, Maryvonne L, Le Dréau G, Gwenvaël LD, Guillonneau X, Xavier G, Lelièvre E, Cécile LE, Slembrouck A, Amélie S, Goureau O, Olivier G, Martinerie C, Cécile M, Marx M, Maria M. Temporal and spatial expression of CCN3 during retina development. Dev Neurobiol 2012; 72:1363-75. [PMID: 22038708 DOI: 10.1002/dneu.20994] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 10/20/2011] [Indexed: 12/21/2022]
Abstract
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.
Collapse
Affiliation(s)
- Maryvonne Laurent
- INSERM UMR S938 Centre de Recherche de Saint-Antoine, Hôpital Saint Antoine Paris F 75012, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Defining the earliest transcriptional steps of chondrogenic progenitor specification during the formation of the digits in the embryonic limb. PLoS One 2011; 6:e24546. [PMID: 21931747 PMCID: PMC3172225 DOI: 10.1371/journal.pone.0024546] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/12/2011] [Indexed: 12/12/2022] Open
Abstract
The characterization of genes involved in the formation of cartilage is of key importance to improve cell-based cartilage regenerative therapies. Here, we have developed a suitable experimental model to identify precocious chondrogenic events in vivo by inducing an ectopic digit in the developing embryo. In this model, only 12 hr after the implantation of a Tgfβ bead, in the absence of increased cell proliferation, cartilage forms in undifferentiated interdigital mesoderm and in the course of development, becomes a structurally and morphologically normal digit. Systematic quantitative PCR expression analysis, together with other experimental approaches allowed us to establish 3 successive periods preceding the formation of cartilage. The “pre-condensation stage”, occurring within the first 3 hr of treatment, is characterized by the activation of connective tissue identity transcriptional factors (such as Sox9 and Scleraxis) and secreted factors (such as Activin A and the matricellular proteins CCN-1 and CCN-2) and the downregulation of the galectin CG-8. Next, the “condensation stage” is characterized by intense activation of Smad 1/5/8 BMP-signaling and increased expression of extracellular matrix components. During this period, the CCN matricellular proteins promote the expression of extracellular matrix and cell adhesion components. The third period, designated the “pre-cartilage period”, precedes the formation of molecularly identifiable cartilage by 2–3 hr and is characterized by the intensification of Sox 9 gene expression, along with the stimulation of other pro-chondrogenic transcription factors, such as HifIa. In summary, this work establishes a temporal hierarchy in the regulation of pro-chondrogenic genes preceding cartilage differentiation and provides new insights into the relative roles of secreted factors and cytoskeletal regulators that direct the first steps of this process in vivo.
Collapse
|
16
|
Janune D, Kubota S, Nishida T, Kawaki H, Perbal B, Iida S, Takigawa M. Novel effects of CCN3 that may direct the differentiation of chondrocytes. FEBS Lett 2011; 585:3033-40. [PMID: 21871891 DOI: 10.1016/j.febslet.2011.08.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/22/2011] [Accepted: 08/10/2011] [Indexed: 10/17/2022]
Abstract
Identification and characterization of local molecules directing the differentiation of chondrocytes to either transient or permanent cartilage are major issues in cartilage biology. Here, we found CCN family protein 3 (CCN3) was abundantly produced in rat developing epiphyseal cartilage. Evaluations in vitro showed that CCN3 repressed epiphyseal chondrocyte proliferation, while promoting matrix production in multiple assays performed. Furthermore, CCN3 enhanced the articular chondrocytic phenotype; whereas it repressed the one representing endochondral ossification. Additionally, the phenotype of growth plate chondrocytes and chondrogenic progenitors also appeared to be affected by CCN3 in a similar manner. These findings suggest a significant role of CCN3 in inducing chondrocytes to articular ones during joint formation.
Collapse
Affiliation(s)
- Danilo Janune
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Kita-ku, Okayama, Japan
| | | | | | | | | | | | | |
Collapse
|
17
|
Kular L, Pakradouni J, Kitabgi P, Laurent M, Martinerie C. The CCN family: A new class of inflammation modulators? Biochimie 2011; 93:377-88. [DOI: 10.1016/j.biochi.2010.11.010] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 11/24/2010] [Indexed: 01/12/2023]
|
18
|
Tryfonidou MA, Lunstrum GP, Hendriks K, Riemers FM, Wubbolts R, Hazewinkel H, Degnin CR, Horton WA. Novel type II collagen reporter mice: New tool for assessing collagen 2α1 expression in vivo and in vitro. Dev Dyn 2011; 240:663-73. [DOI: 10.1002/dvdy.22569] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2011] [Indexed: 12/21/2022] Open
|
19
|
Le Dréau G, Kular L, Nicot AB, Calmel C, Melik-Parsadaniantz S, Kitabgi P, Laurent M, Martinerie C. NOV/CCN3 upregulates CCL2 and CXCL1 expression in astrocytes through beta1 and beta5 integrins. Glia 2010; 58:1510-21. [PMID: 20648642 DOI: 10.1002/glia.21025] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Increasing evidence suggests that CCN matricellular proteins play important roles in inflammation. One of the major cell types that handle inflammation in the brain is the astrocyte, which, upon activation, dramatically increases its production of cytokines and chemokines. Here, we report that NOV/CCN3, added to primary cultured rat brain astrocytes, markedly increased the expression of CCL2 and CXCL1 chemokines, as indicated by ELISA and RT-qPCR assays. This effect was selective, as the production of thirteen other cytokines and chemokines was not affected by NOV. NOV expression by astrocytes was demonstrated by immunocytochemistry and Western blot analysis, and astrocyte transfection with NOV small interfering RNA (siRNA) markedly decreased CXCL1 and CCL2 production, indicating that endogenous NOV played a major role in the control of astrocytic chemokine synthesis. NOV was shown to mediate several of its actions through integrins. Here, we observed that siRNAs against integrins beta1 and beta5 decreased basal and abrogated NOV-stimulated astrocyte expression of CCL2 and CXCL1, respectively. Using a panel of kinase inhibitors, we demonstrated that NOV action on CCL2 and CXCL1 production involved a Rho/ROCK/JNK/NF-kappaB and a Rho/qROCK/p38/NF-kappaB pathway, respectively. Thus, distinct integrins and signaling mechanisms are involved in NOV-induced production of CCL2 and CXCL1 in astrocytes. Finally, astrocytic expression of NOV was detected in rat brain tissue sections, and NOV intracerebral injection increased CCL2 and CXCL1 brain levels in vivo. Altogether, our data shed light on the signaling pathways operated by NOV and strongly suggest that NOV mediates astrocyte activation and, therefore, might play a role in neuroinflammation.
Collapse
Affiliation(s)
- G Le Dréau
- INSERM, UMR_S 938, Centre de Recherche de Saint-Antoine, Hôpital Saint-Antoine, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Canalis E, Smerdel-Ramoya A, Durant D, Economides AN, Beamer WG, Zanotti S. Nephroblastoma overexpressed (Nov) inactivation sensitizes osteoblasts to bone morphogenetic protein-2, but nov is dispensable for skeletal homeostasis. Endocrinology 2010; 151:221-33. [PMID: 19934377 PMCID: PMC2803142 DOI: 10.1210/en.2009-0574] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Overexpression of nephroblastoma overexpressed (Nov), a member of the Cyr 61, connective tissue growth factor, Nov family of proteins, inhibits osteoblastogenesis and causes osteopenia. The consequences of Nov inactivation on osteoblastogenesis and the postnatal skeleton are not known. To study the function of Nov, we inactivated Nov by homologous recombination. Nov null mice were maintained in a C57BL/6 genetic background after the removal of the neomycin selection cassette and compared with wild-type controls of identical genetic composition. Nov null mice were identified by genotyping and absent Nov mRNA in calvarial extracts and osteoblast cultures. Nov null mice did not exhibit developmental skeletal abnormalities or postnatal changes in weight, femoral length, body fat, or bone mineral density and appeared normal. Bone volume and trabecular number were decreased only in 1-month-old female mice. In older mice, after 7 months of age, osteoblast surface and bone formation were increased in females, and osteoclast and eroded surfaces were increased in male Nov null mice. Calvarial osteoblasts from Nov null mice displayed enhanced alkaline phosphatase activity, alkaline phosphatase mRNA, and transactivation of a bone morphogenetic protein (BMP)/phosphorylated mothers against decapentaplegic reporter construct in response to BMP-2. Similar results were obtained after the down-regulation of Nov by RNA interference in ST-2 stromal and MC3T3 cells. Osteoclast number was increased in marrow stromal cell cultures from Nov null mice. Surface plasmon resonance demonstrated direct interactions between Nov and BMP-2. In conclusion, Nov sensitizes osteoblasts to BMP-2, but Nov is dispensable for the maintenance of bone mass.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Research, Saint Francis Hospital and Medical Center, 114 Woodland Street, Hartford, Connecticut 06105-1299, USA.
| | | | | | | | | | | |
Collapse
|
21
|
NOV/CCN3 promotes maturation of cerebellar granule neuron precursors. Mol Cell Neurosci 2009; 43:60-71. [PMID: 19286457 DOI: 10.1016/j.mcn.2009.02.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 02/09/2009] [Accepted: 02/26/2009] [Indexed: 12/20/2022] Open
Abstract
A body of evidence points to the matricial CCN proteins as key regulators of organogenesis. NOV/CCN3, a founder CCN member, is expressed in the developing central nervous system but its functions during neural development have not been studied yet. Here we describe the pattern of NOV expression during rat cerebellar postnatal development and show that NOV expression increases during the second postnatal week, a critical period for the maturation of granule neuron precursors (GNP). NOV transcripts are specifically produced by Purkinje neurons and NOV protein localises extracellularly in the molecular layer and the inner part of the external granule layer, at a key position to control GNP proliferation and migration. In vitro, NOV reduces Sonic Hedgehog-induced GNP proliferation through beta3 integrins and stimulation of GSK3-beta activity whereas NOV stimulates GNP migration through distinct RGD-dependent integrins. These findings identify a new paracrine role of NOV in the development of cerebellar granule neurons.
Collapse
|
22
|
Cooperative regulation of chondrocyte differentiation by CCN2 and CCN3 shown by a comprehensive analysis of the CCN family proteins in cartilage. J Bone Miner Res 2008; 23:1751-64. [PMID: 18597638 PMCID: PMC6956620 DOI: 10.1359/jbmr.080615] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CCN2 is best known as a promoter of chondrocyte differentiation among the CCN family members, and its null mice display skeletal dysmorphisms. However, little is known concerning roles of the other CCN members in chondrocytes. Using both in vivo and in vitro approaches, we conducted a comparative analysis of CCN2-null and wildtype mice to study the roles of CCN2 and the other CCN proteins in cartilage development. Immunohistochemistry was used to evaluate the localization of CCN proteins and other chondrocyte-associated molecules in the two types of mice. Moreover, gene expression levels and the effects of exogenous CCN proteins on chondrocyte proliferation, differentiation, and the expression of chondrocyte-associated genes in their primary chondrocytes were evaluated. Ccn3 was dramatically upregulated in CCN2-null cartilage and chondrocytes. This upregulation was associated with diminished cell proliferation and delayed differentiation. Consistent with the in vivo findings, CCN2 deletion entirely retarded chondrocyte terminal differentiation and decreased the expression of several chondrocyte-associated genes in vitro, whereas Ccn3 expression drastically increased. In contrast, the addition of exogenous CCN2 promoted differentiation strongly and induced the expression of the associated genes, whereas decreasing the Ccn3 expression. These findings collectively indicate that CCN2 induces chondrocyte differentiation by regulating the expression of chondrocyte-associated genes but that these effects are counteracted by CCN3. The lack of CCN2 caused upregulation of CCN3 in CCN2-null mice, which resulted in the observed phenotypes, such as the resultant delay of terminal differentiation. The involvement of the PTHrP-Ihh loop in the regulation of CCN3 expression is also suggested.
Collapse
|
23
|
Inkson CA, Ono M, Kuznetsov SA, Fisher LW, Robey PG, Young MF. TGF-beta1 and WISP-1/CCN-4 can regulate each other's activity to cooperatively control osteoblast function. J Cell Biochem 2008; 104:1865-78. [PMID: 18404666 DOI: 10.1002/jcb.21754] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Wnt-induced secreted protein-1 (WISP-1), like other members of the CCN family, is expressed in skeletal tissues. Its mechanism of action remains unknown. Expression of WISP-1 was analyzed in human bone marrow stroma cells (hBMSC) by RT-PCR. We identified two major transcripts corresponding to those of full-length WISP-1, and of the splice variant WISP-1va which lacks a putative BMP/TGF-beta binding site. To investigate the function of WISP-1 in bone, hBMSC cultures were treated with recombinant human (rh)WISP-1 and analyzed for proliferation and osteogenic differentiation. WISP-1 treatment increased both BrdU incorporation and alkaline phosphatase (AP) activity. Considering the known functional synergy found between the TGF-beta super-family and members of the CCN family, we next tested the effect of WISP-1 on TGF-beta1 activity. We found that rhWISP-1 could reduce rhTGF-beta1 induced BrdU incorporation. Similarly, rhTGF-beta1 inhibited rhWISP-1 induction of AP activity. To explore functional differences between the WISP-1 variants, WISP-1 or WISP-1va were transfected into hBMSC. Both variants could strongly induce BrdU incorporation. However, there were no effects of either variant on AP activity without an additional osteogenic stimulus such as TGF-beta1. Taken together our results suggest a functional relationship between WISP-1 and TGF-beta1. To further define this relationship we analyzed the effect of WISP-1 on TGF-beta signaling. rhWISP-1 significantly reduced TGF-beta1 induced phosphorylation of Smad-2. Our data indicates that full-length WISP-1 and its variant WISP-1va are modulators of proliferation and osteogenic differentiation, and may be novel regulators of TGF-beta1 signaling in osteoblast-like cells.
Collapse
Affiliation(s)
- Colette A Inkson
- Craniofacial and Skeletal Diseases Branch, National Institutes of Craniofacial and Dental Research, National Institutes of Heath, DHHS, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
24
|
Yamaguchi A, Sakamoto K, Minamizato T, Katsube K, Nakanishi S. Regulation of osteoblast differentiation mediated by BMP, Notch, and CCN3/NOV. JAPANESE DENTAL SCIENCE REVIEW 2008. [DOI: 10.1016/j.jdsr.2007.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
25
|
Heath E, Tahri D, Andermarcher E, Schofield P, Fleming S, Boulter CA. Abnormal skeletal and cardiac development, cardiomyopathy, muscle atrophy and cataracts in mice with a targeted disruption of the Nov (Ccn3) gene. BMC DEVELOPMENTAL BIOLOGY 2008; 8:18. [PMID: 18289368 PMCID: PMC2275724 DOI: 10.1186/1471-213x-8-18] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Accepted: 02/20/2008] [Indexed: 02/03/2023]
Abstract
BACKGROUND Signals from the extracellular environment control many aspects of cell behaviour including proliferation, survival, differentiation, adhesion and migration. It is increasingly evident that these signals can be modulated by a group of matricellular proteins called the CCN family. CCN proteins have multiple domains through which they regulate the activities of a variety of signalling molecules including TGFbeta, BMPs and integrins, thereby influencing a wide range of processes in development and disease. Whilst the developmental roles of CCN1 and CCN2 have been elucidated, very little is known about the function of CCN3 (NOV). To investigate this, we have generated mice carrying a targeted mutation in the Nov gene (Novdel3) which reveal for the first time its diverse functions in embryos and adults. RESULTS By replacing Nov exon 3 with a TKneomycin cassette, we have generated Novdel3-/- mice which produce no full length NOV protein and express at a barely detectable level a mutant NOV protein that lacks the VWC domain. In Novdel3-/- embryos, and to a lesser extent in Novdel3+/- embryos, development of the appendicular and axial skeleton was affected with enlarged vertebrae, elongated long bones and digits, delayed ossification, increased bone mineralization and severe joint malformations. Primary embryo fibroblasts from Novdel3-/- mutant embryos showed enhanced chondrogenesis and osteogenesis. Cardiac development was also influenced leading to enlargement and abnormal modelling of the endocardial cushions, associated with septal defects and delayed fusion. In adults, cardiomyopathy was apparent, with hypertrophy and calcification of the septum and left ventricle dilation. Muscle atrophy was seen by 5 months of age, associated with transdifferentiation to fat. Premature tissue degeneration was also seen in the lens, with cataracts present from 6 months. CONCLUSION We have generated the first mice with a mutation in the Nov gene (Novdel3). Our data demonstrate that NOV is a regulator of skeletal and cardiac development, and implicates NOV in various disease processes including cardiomyopathy, muscle atrophy and cataract formation. Novdel3 mutants represent a valuable resource for studying NOV's role in the modulation and co-ordination of multiple signalling pathways that underpin organogenesis and tissue homeostasis.
Collapse
Affiliation(s)
- Emma Heath
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK.
| | | | | | | | | | | |
Collapse
|
26
|
Duong Van Huyen JP, Viltard M, Nehiri T, Freund N, Bélair MF, Martinerie C, Lelongt B, Bruneval P, Lelièvre-Pégorier M. Expression of matrix metalloproteinases MMP-2 and MMP-9 is altered during nephrogenesis in fetuses from diabetic rats. J Transl Med 2007; 87:680-9. [PMID: 17496904 DOI: 10.1038/labinvest.3700562] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Jean-Paul Duong Van Huyen
- INSERM U652, IFR 58, Centre de recherche des Cordeliers, Université René Descartes (Paris 5), Paris, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Rydziel S, Stadmeyer L, Zanotti S, Durant D, Smerdel-Ramoya A, Canalis E. Nephroblastoma overexpressed (Nov) inhibits osteoblastogenesis and causes osteopenia. J Biol Chem 2007; 282:19762-72. [PMID: 17500060 DOI: 10.1074/jbc.m700212200] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nephroblastoma overexpressed (Nov), a member of the Cyr 61, connective tissue growth factor, Nov (CCN) family of proteins, is expressed by osteoblasts, but its function in cells of the osteoblastic lineage is not known. We investigated the effects of Nov overexpression by transducing murine ST-2 stromal and MC3T3 osteoblastic cells with a retroviral vector where Nov is under the control of the cytomegalovirus promoter. We also examined the skeletal phenotype of transgenic mice expressing Nov under the control of the human osteocalcin promoter. Overexpression of Nov in ST-2 cells inhibited the appearance of mineralized nodules and decreased alkaline phosphatase activity and osteocalcin mRNA levels. Nov overexpression inhibited the effect of bone morphogenetic protein (BMP)-2 on the phosphorylation of Smad 1/5/8; on the transactivation of 12xSBE-Oc-pGL3, a BMP/Smad signaling reporter construct, and of Wnt 3 on cytoplasmic beta-catenin levels; and on the transactivation of the Wnt/beta-catenin signaling reporter construct 16xTCF-Luc. Nov overexpression did not activate Notch or transforming growth factor beta signaling. Glutathione S-transferase pulldown assays demonstrated direct Nov-BMP interactions. Nov transgenic mice exhibited osteopenia. In conclusion, Nov binds BMP-2 and antagonizes BMP-2 and Wnt activity, and its overexpression inhibits osteoblastogenesis and causes osteopenia.
Collapse
Affiliation(s)
- Sheila Rydziel
- Department of Research, Saint Francis Hospital and Medical Center, Hartford, Connecticut 06105, USA
| | | | | | | | | | | |
Collapse
|
28
|
Minamizato T, Sakamoto K, Liu T, Kokubo H, Katsube KI, Perbal B, Nakamura S, Yamaguchi A. CCN3/NOV inhibits BMP-2-induced osteoblast differentiation by interacting with BMP and Notch signaling pathways. Biochem Biophys Res Commun 2007; 354:567-73. [PMID: 17250806 DOI: 10.1016/j.bbrc.2007.01.029] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2006] [Accepted: 01/05/2007] [Indexed: 10/23/2022]
Abstract
We elucidate the role of CCN3/NOV, a member of the CCN family proteins, in osteoblast differentiation using MC3T3-E1 osteoblastic cells. Transduction with CCN3 adenovirus (AdCCN3) alone induced no apparent changes in the expression of osteoblast-related markers, whereas cotransduction with BMP-2 adenovirus (AdBMP-2) and AdCCN3 significantly inhibited the AdBMP-2-induced mRNA expression of Runx2, osterix, ALP, and osteocalcin. Immunoprecipitation-western analysis revealed that CCN3 associated with BMP-2. Compared to transduction with AdBMP-2 alone, cotransduction with AdBMP-2 and AdCCN3 attenuated the expression of phosphorylated Smad1/5/8 and the mRNA for Id1, Id2, and Id3. Transduction with AdCCN3 stimulated the expression of cleaved Notch1, the mRNA expression of Hes1 and Hey1/Hesr1, and the promoter activities of Hes1 and Hey1. The inhibitory effects of CCN3 on the expression of BMP-2-induced osteoblast-related markers were nullified in Hey1-deficient osteoblastic cells. These results indicate that CCN3 exerts inhibitory effects on BMP-2-induced osteoblast differentiation by its involvement of the BMP and Notch signaling pathways.
Collapse
Affiliation(s)
- Tokutaro Minamizato
- Section of Oral Pathology, Graduate School of Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8549, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Our bones mostly develop through a process called endochondral ossification. This process is initiated in the cartilage prototype of each bone and continues through embryonic and postnatal development until the end of skeletal growth. Therefore, the central regulator of endochondral ossification is the director of body construction, which is, in other words, the determinant of skeletal size and shape. We suggest that CCN2/CTGF/Hcs24 (CCN2) is a molecule that conducts all of the procedures of endochondral ossification. CCN2, a member of the CCN family of novel modulator proteins, displays multiple functions by manipulating the local information network, using its conserved modules as an interface with a variety of other biomolecules. Under a precisely designed four-dimensional genetic program, CCN2 is produced from a limited population of chondrocytes and acts on all of the mesenchymal cells inside the bone callus to promote the integrated growth of the bone. Furthermore, the utility of CCN2 as regenerative therapeutics against connective tissue disorders, such as bone and cartilage defects and osteoarthritis, has been suggested. Over the years, the pathological action of CCN2 has been suggested. Nevertheless, it can also be regarded as another aspect of the physiological and regenerative function of CCN2, which is discussed as well.
Collapse
Affiliation(s)
- Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | | |
Collapse
|
30
|
Leask A, Abraham DJ. All in the CCN family: essential matricellular signaling modulators emerge from the bunker. J Cell Sci 2006; 119:4803-10. [PMID: 17130294 DOI: 10.1242/jcs.03270] [Citation(s) in RCA: 521] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The CCN family is a group of six secreted proteins that specifically associate with the extracellular matrix. Structurally, CCN proteins are modular, containing up to four distinct functional domains. CCN family members are induced by growth factors and cytokines such as TGFβ and endothelin 1 and cellular stress such as hypoxia, and are overexpressed in pathological conditions that affect connective tissues, including scarring, fibrosis and cancer. Although CCN family members were discovered over a decade ago, the precise biological role, mechanism of action and physiological function of these proteins has remained elusive until recently, when several key mechanistic insights into the CCN family emerged. The CCNs have been shown to have key roles as matricellular proteins, serving as adaptor molecules connecting the cell surface and extracellular matrix (ECM). Although they appear not to have specific high-affinity receptors, they signal through integrins and proteoglycans. Furthermore, in addition to having inherent adhesive abilities that modulate focal adhesions and control cell attachment and migration, they execute their functions by modulating the activity of a variety of different growth factors, such as TGFβ. CCN proteins not only regulate crucial biological processes including cell differentiation, proliferation, adhesion, migration, apoptosis, ECM production, chondrogenesis and angiogenesis, but also have more sinister roles promoting conditions such as fibrogenesis.
Collapse
Affiliation(s)
- Andrew Leask
- CIHR Group in Skeletal Development and Remodeling, Division of Oral Biology, and Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada.
| | | |
Collapse
|
31
|
Abstract
MicroRNAs (miRNAs) are a group of recently discovered small RNAs produced by the cell using a unique process, involving RNA polymerase II, Microprocessor protein complex, and the RNAase III/Dicer endonuclease complex, and subsequently sequestered in an miRNA ribonucleoprotein complex. The biological functions of miRNAs depend on their ability to silence gene expression, primarily via degradation of the target mRNA and/or translational suppression, mediated by the RNA-induced silencing complex (RISC). First discovered in Caenorhabditis elegans (lin-4), miRNAs have now been identified in a wide array of organisms, including plants, zebrafish, Drosophila, and mammals. The expression of miRNAs in multicellular organisms exhibits spatiotemporal, and tissue- and cell-specificity, suggesting their involvement in tissue morphogenesis and cell differentiation. More than 200 miRNAs have been identified or predicted in mammalian cells. Recent studies have demonstrated the importance of miRNAs in embryonic stem cell differentiation, limb development, adipogenesis, myogenesis, angiogenesis and hematopoiesis, neurogenesis, and epithelial morphogenesis. Overexpression (gain-of-function) and inactivation (loss-of-function) are currently the primary approaches to studying miRNA functions. Another family of small RNAs related to miRNAs is the small interfering RNAs (siRNAs), generated by Dicer from long double-stranded RNAs (dsRNAs), and produced from an induced transgene, a viral intruder, or a rogue genetic element. siRNAs silence genes via either mRNA degradation, using the RISC, or DNA methylation. siRNAs are actively being applied in basic, functional genetic studies, particularly in the generation of gene knockdown animals, as well as in gene knockdown studies of cultured cells. These studies have provided invaluable information on the specific function(s) of individual genes. siRNA technology also presents exciting potential as a therapeutic approach in disease prevention and treatment, as suggested by a recent study targeting apolipoprotein B (ApoB) in primates. Further elucidation of how miRNAs and other small RNAs interact with known and yet-to-be identified gene regulatory pathways in the cell should provide us with a more in-depth understanding of the mechanisms regulating cellular function and differentiation, and facilitate the application of small RNA technology in disease control and treatment.
Collapse
Affiliation(s)
- Lin Song
- Cartilage Biology and Orthopaedics Branch of the National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892, USA
| | | |
Collapse
|