Elsevier

Experimental Cell Research

Volume 315, Issue 14, 15 August 2009, Pages 2386-2398
Experimental Cell Research

Research Article
Smurf2 induces degradation of GSK-3β and upregulates β-catenin in chondrocytes: A potential mechanism for Smurf2-induced degeneration of articular cartilage

https://doi.org/10.1016/j.yexcr.2009.05.019Get rights and content

Abstract

We have previously demonstrated that Smurf2 is highly expressed in human osteoarthritis (OA) tissue, and overexpression of Smurf2 under the control of the type II collagen promoter (Col2a1) induces an OA-like phenotype in aged Col2a1-Smurf2 transgenic mice, suggesting that Smurf2 is located upstream of a signal cascade which initiates OA development. However, the factors downstream of Smurf2 in this signal cascade and how Smurf2-induced OA is initiated are largely unknown. In this study, we further characterized the phenotypic changes in Col2a1-Smurf2 transgenic and WT articular cartilage from the postnatal stage to adulthood. We found that the articular cartilage degeneration occurring at the cartilage surface in 6 month-old Col2a1-Smurf2 transgenic mice progressed from an expanded hypertrophic domain in the basal layer of the deep articular cartilage at 2.5 weeks of age, which may lead to an accelerated calcification and ectopic ossification of this region at 1 month of age, and aggregation and maturation of articular chondrocytes in the middle and deep zones at 2 months and 4.5 months of age, respectively. Furthermore, we discovered that ectopically expressed Smurf2 interacted with GSK-3β and induced its ubiquitination and subsequent proteasomal degradation, and hence upregulated β-catenin in Col2a1-Smurf2 transgenic chondrocytes ex vivo. It is therefore likely that Smurf2-mediated upregulation of β-catenin through induction of proteasomal degradation of GSK-β in chondrocytes may activate articular chondrocyte maturation and associated alteration of gene expression, the early events of OA.

Introduction

The process of endochondral ossification consists of multiple stages. First, mesenchymal cells aggregate to form condensations, which subsequently differentiate into two types of cells: chondrocytes that form cartilage elements and osteoblast lineage cells that form the perichondrium surrounding the cartilage rudiment. Second, the differentiated chondrocytes undergo proliferation to form orderly parallel columns. The proliferating chondrocytes synthesize and deposit cartilage matrix such as type II collagen (Col2), and proteoglycans. Third, chondrocytes exit the cell cycle and mature into hypertrophic chondrocytes, and express the stage-specific marker type X collagen (ColX). Finally, terminally differentiated hypertrophic chondrocytes express matrix metalloproteinase (MMP) 13, and the cartilage matrix is degraded and replaced by bone.

Chondrocyte differentiation and maturation during endochondral ossification are tightly regulated by several key growth factors and transcription factors. Members of the TGF-β superfamily such as TGF-β isoforms and BMPs play important roles in the regulation of chondrocyte differentiation and maturation during this process. For example, TGF-β induces chondrogenesis including chondrocyte differentiation and cartilage matrix synthesis in developing limb buds [1], [2]. On the other hand, TGF-β signaling inhibits chondrocyte maturation and ColX expression during long bone lengthening, evidenced by loss of TGF-β signaling in the growth plate resulting in increased thickness of hypertrophic zone and upregulation of ColX expression [3], [4]. Similar to the stimulatory role of TGF-β signaling in chondrogenesis, BMP signaling is critical for mesenchymal condensation and chondrocyte differentiation during limb bud development [5], [6], [7], [8], [9]. However, in contrast to the inhibitory effect of TGF-β signaling on chondrocyte maturation, BMP signaling promotes this process [6], [10], [11]. TGF-β/BMP signaling is initiated by binding of these ligands to their receptors, which induces receptor phosphorylation and phosphorylation of receptor regulated Smads including Smad1, Smad5, Smad8 (BMP receptor), Smad2, and Smad3 (TGF-β receptor) [12], [13], [14], [15]. Recently, accumulating evidence suggests that canonical Wnt signaling, which is transduced via β-catenin, is implicated in multiple-steps of endochondral bone formation [16], [17], [18], [19], [20]. For example, ectopic β-catenin in cells of the chondrogenic lineage inhibits chondrocyte differentiation but stimulates chondrocyte maturation and ossification during embryonic development [16], [20].

In general, β-catenin levels are upregulated by Wnt ligands. β-catenin levels are normally limited through continuous proteasome-mediated degradation of phosphorylated β-catenin, which is catalyzed by the enzyme GSK-3β in a “destruction complex” brought together by Axin and APC [21], [22]. Upon Wnt ligand binding to its receptors, the complex is disassociated, and the kinase activity of GSK-3β is suppressed, and hence non-phosphorylated β-catenin accumulates in the cytoplasm and translocates into the nucleus to activate expression of Wnt target genes [23]. The β-catenin protein levels are therefore tightly regulated by the Wnt-mediated activity of the “destruction complex”, and any mechanism by which normal β-catenin regulation is uncoupled from Wnt signaling control could result in dysregulation of osteoblast/chondrocyte function, leading to alterations in bone mass or degenerative joints [24], [25], [26]. However, our understanding of the abnormal regulation of β-catenin in these diseases and animal models remains incomplete.

Osteoarthritis (OA) is a degenerative joint disease, and its developmental process recapitulates many of the events that occur during endochondral ossification. The primary feature in OA-affected joints is articular cartilage degeneration. Unlike the cartilage in the developing growth plate, in which chondrocytes mature and matrix turns over rapidly, the articular cartilage is a permanent tissue with minimal turnover of its cells and matrix [27], [28], [29]. The function of articular cartilage that ensures the frictionless movement of joints depends on the unique architecture of the extracellular matrix and zonal articular chondrocytes. The matrix is a highly hydrated network of collagen fibrils embedded in a gel of negatively charged proteoglycan molecules. The articular chondrocytes, which are sparsely distributed in the matrix, are responsible for maintenance of the matrix. Articular cartilage is developed postnatally and cross-link maturation of the collagen network is completed by adulthood. Appearance of articular cartilage is at postnatal 2 weeks when a small domain of secondary ossification occurs in the center of the epiphysis, which separates the articular cartilage from the growth plate cartilage [9], [30]. At this stage, the deep zone of the newly formed articular cartilage is a growth plate-like tissue and contains a thin layer of enlarged chondrocytes expressing ColX, which may be associated with formation of the calcified zone seen in adult articular cartilage [31], [32], [33]. By 1 month of age, the secondary ossification center is expanded, and the zonal articular cartilage formation is completed [33]. Articular chondrocytes normally do not undergo maturation. However, during OA development, articular chondrocytes undergo maturation, and express maturational marker genes such as ColX [34], [35], [36], [37], and MMP-13 [37], [38], [39], [40], which in turn degrades matrix and leads to articular cartilage degeneration [39], [41], [42], [43]. As OA progresses, osteophytes formed via endochondral ossification are often seen at the margins of the joint and bridge the joint cavity. While OA is a leading cause of disability in older people [44], [45], the underlying molecular mechanism is largely unknown. Although loss of TGFβ/Smad3 signaling in skeletal cells via genetic approaches results in OA in mice such as in the dominant-negative TGFβ type II receptor transgenic mice and in the Smad3 KO mice [3], [4], or local administration/overexpression of TGF-β in the knee joints induces osteophyte formation [46], [47], [48], these events do not occur in humans with OA. Recently, several lines of evidence suggest a role for increased Wnt/β-catenin signaling in OA development. For example, mutations in FRZB, a soluble Wnt antagonist, are OA risk factors, and increase hip OA susceptibility in women [26], [49], [50], which are further confirmed by the phenotype of Frzb KO mice, which are more susceptible to chemically-induced OA [51]. Consistently, high β-catenin protein levels were detected in human OA chondrocytes, but not in normal articular cartilage [52], [53], [54], indicating that increased β-catenin signaling in articular chondrocytes may be responsible for a conversion of normal cells to OA cells. However, the upstream signaling pathway(s) that dictates upregulation of β-catenin in articular chondrocytes during OA initiation is not clear.

We have recently demonstrated that Smurf2 is highly expressed in human OA tissue, and that overexpression of Smurf2 under the control of the type II collagen promoter (Col2a1) induces OA in mice, suggesting that Smurf2 is located upstream of a signaling cascade that is responsible for OA initiation in humans [37]. In some cell lines, ectopically overexpressed Smurf2 directly interacts with Smad1 and Smad2, and indirectly interacts with the type I receptor of TGF-β and β-catenin via an adaptor Smad7 for ubiquitination and proteasomal degradation [55], [56], [57], [58]. However, ectopic Smurf2 in Col2a1-Smurf2 transgenic chondrocytes does not induce the proteasomal degradation of these known targets identified in vitro but induces that of exogenous TGF-β activated phosphorylated Smad3, and decreases associated TGF-β/Smad3 signaling ex vivo [37]. Notably, the basal level of phosphorylated Smad3 and TGF-β/Smad3 signaling is slightly decreased in resting Col2a1-Smurf2 transgenic chondrocytes versus resting WT cells [37]. Given that Smad3 KO mice and Col2a1-Smurf2 transgenic mice develop severe OA at a similar age [4], [37], the mild loss of TGF-β/Smad3 signaling in Col2a1-Smurf2 transgenic chondrocytes may not be the major mechanism underlying Smurf2-induced OA in the transgenic mice. In this study, we demonstrate that ectopic Smurf2 in Col2a1-Smurf2 transgenic chondrocytes targets basal GSK-3β for ubiquitination and proteasomal degradation and increases the basal levels of β-catenin protein and signaling within these cells ex vivo. This novel signal cascade may represent a predominant mechanism that activates articular chondrocyte maturation, a critical early event during OA development.

Section snippets

Histology, in situ hybridization, and μ-CT

Newborn limbs and knee joints at various stages were dissected in cold PBS, fixed in 4% paraformaldehyde at 4 °C for 2–3 days, and embedded in paraffin. Five micrometer thick sagittal sections were harvested every 10 μm (limb), 15 μm (2 week-old), or 30 μm (≥ 1 month) from the medial joint compartment of the right knee. Four–six serial sections were used for quantification of cartilage/chondrocyte morphology as we described previously [20], [37]. Specifically, the number of articular

Expansion of ColX-expressing domain in deep articular cartilage of postnatal Col2a1-smurf2 transgenic mice

We have previously shown that ectopically overexpressed Smurf2 in articular chondrocytes induces an OA-like phenotype in Col2a1-Smurf2 transgenic mice, which is characterized by articular cartilage degeneration and osteophyte formation at over 6 months of age [20], [37]. These phenotypic changes could result from earlier abnormalities in transgenic articular cartilage. To test this hypothesis, we first examined the articular cartilage phenotype in WT and transgenic mice at 2.5 weeks of age when

Articular chondrocyte maturation in Col2a1-smurf2 transgenic mice

The OA-like phenotype characterized by degeneration of articular cartilage and periarticular extra bone formation (osteophyte) in knee joints from over 6 month-old Col2a1-Smurf2 transgenic mice may arise from earlier articular cartilage abnormalities. To test this hypothesis, we examined the phenotypic changes in the articular cartilage of WT and Col2a1-Smurf2 transgenic mice at various ages. At 2.5 weeks of age, WT articular cartilage has just separated from growth plate cartilage by a newly

Acknowledgments

This work was supported by NIH/NIAMS RO1 AR045700 (Randy N Rosier), NIH/NIAMS P50 AR054041 (Randy N Rosier), Aircast Foundation (Qiuqian Wu), and NIH/NIAMS R01 AR054465 (Di Chen). We thank Bojian Liang, Erica J Dussmann, and Ryan M Tierney for their technical support. We appreciate the gift of human Smurf2 cDNA from Wrana JL, Ph.D. (University of Toronto, Canada).

References (65)

  • E.V. Tchetina et al.

    Chondrocyte hypertrophy can be induced by a cryptic sequence of type II collagen and is accompanied by the induction of MMP-13 and collagenase activity: implications for development and arthritis

    Matrix Biol.

    (2007)
  • I. Girkontaite et al.

    Immunolocalization of type X collagen in normal fetal and adult osteoarthritic cartilage with monoclonal antibodies

    Matrix Biol.

    (1996)
  • G.E. Kempson et al.

    The tensile properties of the cartilage of human femoral condyles related to the content of collagen and glycosaminoglycans

    Biochim. Biophys. Acta.

    (1973)
  • H.M. van Beuningen et al.

    Osteoarthritis-like changes in the murine knee joint resulting from intra-articular transforming growth factor-beta injections

    Osteoarthr. Cartil.

    (2000)
  • S.J. Kim et al.

    Beta-catenin regulates expression of cyclooxygenase-2 in articular chondrocytes

    Biochem. Biophys. Res. Commun.

    (2002)
  • S.G. Hwang et al.

    Wnt-7a causes loss of differentiated phenotype and inhibits apoptosis of articular chondrocytes via different mechanisms

    J. Biol. Chem.

    (2004)
  • P. Kavsak et al.

    Smad7 binds to Smurf2 to form an E3 ubiquitin ligase that targets the TGF beta receptor for degradation

    Mol. Cell.

    (2000)
  • X. Lin et al.

    Smurf2 is an ubiquitin E3 ligase mediating proteasome-dependent degradation of Smad2 in transforming growth factor-beta signaling

    J. Biol. Chem.

    (2000)
  • G. Han et al.

    Smad7-induced beta-catenin degradation alters epidermal appendage development

    Dev. Cell

    (2006)
  • R. Shen et al.

    Cyclin D1-cdk4 induce runx2 ubiquitination and degradation

    J. Biol. Chem.

    (2006)
  • O. Jacenko et al.

    A dominant interference collagen X mutation disrupts hypertrophic chondrocyte pericellular matrix and glycosaminoglycan and proteoglycan distribution in transgenic mice

    Am. J. Pathol.

    (2001)
  • H.J. Kim et al.

    Collagen/annexin V interactions regulate chondrocyte mineralization

    J. Biol. Chem.

    (2008)
  • W.M. Kulyk et al.

    Promotion of embryonic chick limb cartilage differentiation by transforming growth factor-beta

    Del. Biol.

    (1989)
  • J. Chimal-Monroy et al.

    Differential effects of transforming growth factors beta 1, beta 2, beta 3 and beta 5 on chondrogenesis in mouse limb bud mesenchymal cells

    Int. J. Dev. Biol.

    (1997)
  • R. Serra et al.

    Expression of a truncated, kinase-defective TGF-beta type II receptor in mouse skeletal tissue promotes terminal chondrocyte differentiation and osteoarthritis

    J. Cell. Biol.

    (1997)
  • X. Yang et al.

    TGF-β/Smad3 signals repress chondrocyte hypertrophic differentiation and are required for maintaining articular cartilage

    J. Cell. Biol.

    (2001)
  • H. Zou et al.

    Distinct roles of type I bone morphogenetic protein receptors in the formation and differentiation of cartilage

    Genes Dev.

    (1997)
  • B.S. Yoon et al.

    Bmpr1a and Bmpr1b have overlapping functions and are essential for chondrogenesis in vivo

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • A. Bandyopadhyay et al.

    Genetic analysis of the roles of BMP2, BMP4, and BMP7 in limb patterning and skeletogenesis

    PLoS Genet.

    (2006)
  • E. Minina et al.

    BMP and Ihh/PTHrP signaling interact to coordinate chondrocyte proliferation and differentiation

    Development

    (2001)
  • C.H. Heldin et al.

    TGF-β signaling from cell membrane to nucleus through SMAD proteins

    Nature

    (1997)
  • J. Massaguē et al.

    Transcriptional control by the TGF-β/SMAD signaling system

    EMBO J.

    (2000)
  • Cited by (54)

    • Celecoxib nanocrystal-loaded dissolving microneedles with highly efficient for osteoarthritis treatment

      2022, International Journal of Pharmaceutics
      Citation Excerpt :

      Osteoarthritis (OA) is a degenerative disease that affects a variety of joints, including minor and major joints (Yi et al., 2021; Park et al., 2021). Excessive usage of these joints can result in articular cartilage loss, subchondral bone alterations, joint discomfort and swelling, and other problems (Diab et al., 2017; Wu et al., 2009; Lu et al., 2015). As a result, the primary aims of OA treatment are to reduce patient discomfort and halt disease progression.

    • The functions and regulation of Smurfs in cancers

      2020, Seminars in Cancer Biology
      Citation Excerpt :

      Surprisingly, both wild-type and catalytically inactive Smurf2 stabilize CNKSR2 levels in cancer cells [83]. Smurf2 promotes Wnt signaling by triggering the ubiquitination and degradation of negative regulatory proteins, including GSK3β [84] and AXIN1 [60]. Furthermore, the β-TrCP complex mediates the degradation of β-catenin, while Smurf2 promotes the degradation of β-TrCP [80,85].

    • New insights in osteoarthritis diagnosis and treatment: Nano-strategies for an improved disease management

      2023, Wiley Interdisciplinary Reviews: Nanomedicine and Nanobiotechnology
    View all citing articles on Scopus
    1

    Current address: Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14642, USA.

    View full text