CCN3 (NOV) Drives Degradative Changes in Aging Articular Cartilage

International Journal of Molecular Sciences - Tập 21 Số 20 - Trang 7556
Miho Kuwahara1,2,3, Koichi Kadoya1,4, Sei Kondo1,2, Shanqi Fu1,2, Yoshiko Miyake1,2, Ayako Ogo1,2, Mitsuaki Ono1,5, Takayuki Furumatsu1,6, Eiji Nakata1,6, Takako Sasaki7, Shogo Minagi3, Masaharu Takigawa8, Satoshi Kubota1,2, Takako Hattori1,2
1Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan;
2Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine,
3Department of Occlusal and Oral Functional Rehabilitation, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan;
4Department of Oral and Maxillofacial Surgery, Okayama University Graduate School of Medicine,
5Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine,
6Department of Orthopedic Surgery, Okayama University Graduate School of Medicine,
7Department of Biochemistry, Faculty of Medicine, Oita University, Oita 879-5593, Japan;
8Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Dental School/Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan;

Tóm tắt

Aging is a major risk factor of osteoarthritis, which is characterized by the degeneration of articular cartilage. CCN3, a member of the CCN family, is expressed in cartilage and has various physiological functions during chondrocyte development, differentiation, and regeneration. Here, we examine the role of CCN3 in cartilage maintenance. During aging, the expression of Ccn3 mRNA in mouse primary chondrocytes from knee cartilage increased and showed a positive correlation with p21 and p53 mRNA. Increased accumulation of CCN3 protein was confirmed. To analyze the effects of CCN3 in vitro, either primary cultured human articular chondrocytes or rat chondrosarcoma cell line (RCS) were used. Artificial senescence induced by H2O2 caused a dose-dependent increase in Ccn3 gene and CCN3 protein expression, along with enhanced expression of p21 and p53 mRNA and proteins, as well as SA-β gal activity. Overexpression of CCN3 also enhanced p21 promoter activity via p53. Accordingly, the addition of recombinant CCN3 protein to the culture increased the expression of p21 and p53 mRNAs. We have produced cartilage-specific CCN3-overexpressing transgenic mice, and found degradative changes in knee joints within two months. Inflammatory gene expression was found even in the rib chondrocytes of three-month-old transgenic mice. Similar results were observed in human knee articular chondrocytes from patients at both mRNA and protein levels. These results indicate that CCN3 is a new senescence marker of chondrocytes, and the overexpression of CCN3 in cartilage may in part promote chondrocyte senescence, leading to the degeneration of articular cartilage through the induction of p53 and p21.

Từ khóa


Tài liệu tham khảo

Hermann, 2018, Current Treatment Options for Osteoarthritis, Curr. Rheumatol. Rev., 14, 108, 10.2174/1573397113666170829155149

Ashford, 2014, Osteoarthritis: A review, Nurse Pract., 39, 1, 10.1097/01.NPR.0000445886.71205.c4

Hou, 2018, Cellular senescence in osteoarthritis and anti-aging strategies, Mech. Ageing Dev., 175, 83, 10.1016/j.mad.2018.08.002

Livshits, 2009, Interleukin-6 is a significant predictor of radiographic knee osteoarthritis: The Chingford study, Arthritis Rheum., 60, 2037, 10.1002/art.24598

Philipot, 2014, p16INK4a and its regulator miR-24 link senescence and chondrocyte terminal differentiation-associated matrix remodeling in osteoarthritis, Arthritis Res. Ther., 16, R58, 10.1186/ar4494

Ashraf, 2016, Regulation of senescence associated signaling mechanisms in chondrocytes for cartilage tissue regeneration, Osteoarthr. Cartil., 24, 196, 10.1016/j.joca.2015.07.008

Tsuchida, 2014, Cytokine profiles in the joint depend on pathology, but are different between synovial fluid, cartilage tissue and cultured chondrocytes, Arthritis Res., 16, 441, 10.1186/s13075-014-0441-0

Hui, 2015, Oxidative changes and signalling pathways are pivotal in initiating age-related changes in articular cartilage, Ann. Rheum. Dis., 75, 449, 10.1136/annrheumdis-2014-206295

Wang, 2015, Mitochondrial Biogenesis Is Impaired in Osteoarthritis Chondrocytes but Reversible via Peroxisome Proliferator-Activated Receptor γ Coactivator 1α, Arthritis Rheumatol., 67, 2141, 10.1002/art.39182

Blanco, 2011, The role of mitochondria in osteoarthritis, Nat. Rev. Rheumatol., 7, 161, 10.1038/nrrheum.2010.213

Ulgherait, 2014, AMPK Modulates Tissue and Organismal Aging in a Non-Cell-Autonomous Manner, Cell Rep., 8, 1767, 10.1016/j.celrep.2014.08.006

Olmer, 2015, The relationship of autophagy defects to cartilage damage during joint aging in a mouse model, Arthritis Rheumatol., 67, 1568, 10.1002/art.39073

Taniguchi, 2010, Autophagy is a protective mechanism in normal cartilage, and its aging-related loss is linked with cell death and osteoarthritis, Arthritis Rheum., 62, 791, 10.1002/art.27305

Verzijl, 2002, Crosslinking by advanced glycation end products increases the stiffness of the collagen network in human articular cartilage: A possible mechanism through which age is a risk factor for osteoarthritis, Arthritis Rheum., 46, 114, 10.1002/1529-0131(200201)46:1<114::AID-ART10025>3.0.CO;2-P

Holbourn, 2008, The CCN family of proteins: Structure–function relationships, Trends Biochem. Sci., 33, 461, 10.1016/j.tibs.2008.07.006

Takigawa, 2016, The CCN Proteins: An Overview, Adv. Struct. Saf. Stud., 1489, 1

Leask, 2006, All in the CCN family: Essential matricellular signaling modulators emerge from the bunker, J. Cell Sci., 119, 4803, 10.1242/jcs.03270

Kawaki, 2008, 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., 23, 1751, 10.1359/jbmr.080615

Eisenberg, 2013, Human housekeeping genes, revisited, Trends Genet., 29, 569, 10.1016/j.tig.2013.05.010

Rubingh, 2019, Identification of stable senescence-associated reference genes, Aging Cell, 18, e12911, 10.1111/acel.12911

Chambers, 2002, Expression of collagen and aggrecan genes in normal and osteoarthritic murine knee joints, Osteoarthr. Cartil., 10, 51, 10.1053/joca.2001.0481

Loeser, 2012, Microarray analysis reveals age-related differences in gene expression during the development of osteoarthritis in mice, Arthritis Rheum., 3, 705, 10.1002/art.33388

Childs, 2015, Cellular senescence in aging and age-related disease: From mechanisms to therapy, Nat. Med., 21, 1424, 10.1038/nm.4000

Desprez, 2010, The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression, Annu. Rev. Pathol. Mech. Dis., 5, 99, 10.1146/annurev-pathol-121808-102144

Serrano, 2014, Cellular senescence: From physiology to pathology, Nat. Rev. Mol. Cell Biol., 15, 482, 10.1038/nrm3823

Aigner, 1997, Phenotypic modulation of chondrocytes as a potential therapeutic target in osteoarthritis: A hypothesis, Ann. Rheum. Dis., 56, 287, 10.1136/ard.56.5.287

Troeberg, 2012, Proteases involved in cartilage matrix degradation in osteoarthritis, Biochim. Biophys. Acta., 1824, 133, 10.1016/j.bbapap.2011.06.020

Kamekura, 2005, Osteoarthritis development in novel experimental mouse models induced by knee joint instability, Osteoarthr. Cartil., 13, 632, 10.1016/j.joca.2005.03.004

Shimoyama, 2010, CCN3 Inhibits Neointimal Hyperplasia Through Modulation of Smooth Muscle Cell Growth and Migration, Arter. Thromb. Vasc. Biol., 30, 675, 10.1161/ATVBAHA.110.203356

Heath, E., Tahri, D., Andermarcher, E., Schofield, P.N., Fleming, S., and Boulter, C.A. (2008). Abnormal skeletal and cardiac development, cardiomyopathy, muscle atrophy and cataracts in mice with a targeted disruption of the Nov (Ccn3) gene. BMC Dev. Biol., 8.

Roddy, 2015, Targeted mutation of NOV/CCN3 in mice disrupts joint homeostasis and causes osteoarthritis-like disease, Osteoarthr. Cartil., 23, 607, 10.1016/j.joca.2014.12.012

Suresh, 2013, The matricellular protein CCN3 regulates NOTCH1 signalling in chronic myeloid leukaemia, J. Pathol., 231, 378, 10.1002/path.4246

He, 2017, Senescence in Health and Disease, Cell, 169, 1000, 10.1016/j.cell.2017.05.015

Joliot, 1992, Proviral rearrangements and overexpression of a new cellular gene (nov) in myeloblastosis-associated virus type 1-induced nephroblastomas, Mol. Cell. Biol., 12, 10

Chevalier, 1998, novH: Differential expression in developing kidney and Wilm’s tumors, Am. J. Pathol., 152, 1563

Yu, 2003, NOV (CCN3) regulation in the growth plate and CCN family member expression in cartilage neoplasia, J. Pathol., 201, 609, 10.1002/path.1468

Akashi, 2017, Metabolic regulation of the CCN family genes by glycolysis in chondrocytes, J. Cell Commun. Signal., 12, 245, 10.1007/s12079-017-0420-8

Huang, 2019, NOV/CCN3 induces cartilage protection by inhibiting PI3K/AKT/mTOR pathway, J. Cell. Mol. Med., 23, 7525, 10.1111/jcmm.14621

Peidl, 2018, A friend in knee: CCN3 may inhibit osteoarthritis progression, J. Cell. Commun. Signal., 12, 489, 10.1007/s12079-017-0446-y

Hattori, 2010, SOX9 is a major negative regulator of cartilage vascularization, bone marrow formation and endochondral ossification, Development, 137, 901, 10.1242/dev.045203

Tomita, N., Hattori, T., Itoh, S., Aoyama, E., Yao, M., Yamashiro, T., and Takigawa, M. (2013). Cartilage-specific over-expression of CCN family member 2/connective tissue growth factor (CCN2/CTGF) stimulates insulin-like growth factor expression and bone growth. PLoS ONE, 8.

King, 2003, The establishment and characterization of an immortal cell line with a stable chondrocytic phenotype, J. Cell. Biochem., 89, 992, 10.1002/jcb.10571

Itahana, 2013, Colorimetric detection of senescence-associated β galactosidase, Methods. Mol. Biol., 965, 143, 10.1007/978-1-62703-239-1_8