Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and TGFβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This fundamental work advances our understanding of the role of Cbfβ in maintaining articular cartilage homeostasis and the underlying mechanisms. The evidence supporting the conclusion is mostly convincing, although including additional experiments and discussions would have strengthened the study. This paper is of potential interest to skeletal biologists and orthopaedic surgeons who study the pathogenesis and the therapeutics of osteoarthritis.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that Cbfβ, (subunit of a heterodimeric Cbfβ/Runx1,Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfβ in tamoxifen-induced Cbfβ f/f Col2α1-CreER T mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/YAP signaling and TGF-β signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfβ overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfβ overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfβ may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and TGFβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfβ overexpression could be an effective strategy for treatment of OA.

Article activity feed

  1. Author Response

    Reviewer #2 (Public Review):

    In this manuscript, Chen et al. reported that the core binding factor beta (Cbfβ), a heterodimeric subunit of the RUNX family transcription factors (TFs), is crucial in maintaining cartilage homeostasis and counteracting traumatic OA pathology. Using mouse models in which Cbfβ is conditionally inactivated in the Col2a1+ and Acan+ cells, the authors claimed that Cbfβ ablation led to articular cartilage (AC) degeneration, which is associated with aberrant cartilage gene expression and chondrocyte signaling, particularly the elevated Wnt/Catenin and the decreased Hippo/YAP and TGFβ signaling. The authors further showed that Cbfβ transcripts are decreased in human OA cartilage, and sustaining Cbfβ expression in mouse knee joints mitigated the severity of surgery-evoked OA.

    On the whole, the work reported is interesting and exciting. Genetic and biochemical data support key statements. Both in vivo and in vitro experiments were well designed with proper controls; semiquantitative data were digitalized and processed for statistical significance. Furthermore, new findings were adequately discussed in contrast to the current available knowledge. However, the conceptual novelty of this study is slightly compromised by recent publications showing that Cbfβ reduction is associated with OA (Che et al. 2023; Li et al. 2021). Also, the authors claimed that multiple signaling pathways were affected by Cbfβ ablation in cartilage cells; many of them, however, are indirect effects given the nature of Cbfβ as a TF. The authors also showed that pSMAD2/3 and active βCatenin decreased and increased upon Cbfβ depletion in the mouse AC cartilage. However, how the deficiency of Cbfβ, a widely expressed TF, affected the posttranslational modification of SMAD2/3 and βCatenin is unclear and needs further discussion. Overall, Cbfβ's role in cartilage and OA pathology is an emerging area of study; the authors provided a set of genetic evidences showing that Cbfβ is indispensable for cartilage homeostasis.

    We thank the reviewer for the positive appraisal of our manuscript. We greatly appreciate the insightful comments and critiques. In accordance with the reviewer’s suggestions, we have thoroughly revised all parts of the manuscript. We are glad that the reviewers considered our work to be of interest, and we are grateful for this opportunity to resubmit our manuscript. With regard to concerns of novelty of our study, Li et al’s study only reported the relationship between abnormal Cbfβ expression in human cartilage and osteoarthritis. Che et al’s study employed Cbfβf/fAggrecan-cre mice, while our study used a novel inducible Cbfβf/fCol2α1CreERT mouse model. While the Aggrecan-creERT system provides valuable insights into the role of Cbfβ in differentiated cartilage cells and its implications in the advanced stages of osteoarthritis, our current study also used Cbfβf/fCol2α1-CreERT aimed to explore the gene's function from a broader perspective. Previous study points out that Col2α1 is expressed in both early and late stage of chondrogenesis, including skeletal mesenchymal cells, perichondrium and presumptive joint cells, but aggrecan is expressed specifically in differentiated chondrocytes(1). However, studies show that not only differentiated chondrocytes but also chondrocyte progenitors are involved in OA pathogenesis(2). In our current study, the Col2α1-CreERT system allowed us to investigate Cbfβ's role not only in mature chondrocytes but also in early chondroprogenitor cells, offering a comprehensive view of Cbfβ’s involvement in cartilage in osteoarthritis. Therefore, the use of the Cbfβf/fCol2α1-CreERT mouse mutant strain was instrumental in expanding our understanding of Cbfβ's multifaceted role in osteoarthritis, highlighting its importance not only in mature cartilage but also in the early stages of cartilage formation and differentiation. In addition to the different types of Cre used compared to our previous study, our current study also used gain-of-function approach in ACLT-induced OA disease model to understand the potential therapeutic function of Cbfβ in OA pathological condition. Adding our current findings to our previous research, we can now piece together a more complete picture of Cbfβ's role across the entire spectrum of cartilage development in osteoarthritis.

    We agree with the reviewer that how the deficiency of Cbfβ, a widely expressed TF, affected the posttranslational modification of SMAD2/3 and βCatenin is unclear and needs further exploration. So far there is no clear explanation of this, which is why we used RNA-seq and heatmap analysis to examine other genes expression which could help to uncover the mechanism underlying these results. Interestingly, Che et al’s result showed that TGFB signaling (P-Smad3) increased in Cbfβf/fAggrecan-cre mice, while our data showed that TGFB signaling (both PSmad3 and Smad3) decreased in Cbfβf/fCol2α1-CreERT mice as shown in our results in Figure 8. These results were also confirmed by RNA-seq analysis as shown in the heatmaps in figure 5.

    These differences could be the result of different mouse ages used in our study and Che et al’s study.

    1. Blaney Davidson EN, van de Loo FA, van den Berg WB, van der Kraan PM. How to build an inducible cartilagespecific transgenic mouse. Arthritis Res Ther. 2014;16(3):210.

    2. Tong L, Yu H, Huang X, Shen J, Xiao G, Chen L, et al. Current understanding of osteoarthritis pathogenesis and relevant new approaches. Bone Res. 2022;10(1):60.

    Reviewer #3 (Public Review):

    The authors comprehensively demonstrated the Cbfβ gene, which is involved in articular cartilage homeostasis, can promote articular cartilage regeneration and repair in osteoarthritis (OA) through regulating Hippo/YAP signaling TGF-β signaling, and canonical Wnt signaling. First, the authors demonstrated the deletion of Cbfβ can induce the OA phenotypes including decreased articular cartilage and osteoblasts, and increased osteoclasts and subchondral bone hyperplasia, and induce the early onset of OA. Additionally, the authors showed that the deficiency of Cbfβ in cartilage can increase canonical Wnt signaling and decrease TGF-β and Hippo signaling. Finally, the authors demonstrated that the overexpression of Cbfβ can inhibit Wnt signaling and enhance Hippo/YAP signaling in knee joints articular cartilage of ACLT-induced OA mice and protect against ACLT-induced OA. The manuscript is overall well-constructed, and the authors provided evidence to support their findings.

    In Fig. 7I, it could be better to show the statistical analysis between normal and AAV-mediated Cbfβ ACLT mice groups.

    We thank the reviewer for bringing this to our attention. In the revised figure 7I, we have included the statistical analysis between normal and AAV-mediated Cbfβ ACLT mice groups.

    In Fig. 9H-K, in the quantification analysis, the OARSI score in the DMM+AAV-YFP group is higher than in the sham group significantly. However, the SO staining results appear to show no significant difference between the DMM+AAV-luc-YFP group (Fig. 9I) and the sham group (Fig. 9H).

    We thank the reviewer for bringing this to our attention. Although both the sham and DMM+AAV-luc-YFP group stain positive for SO, the SO stain intensity of the DMM+AAV-lucYFP group is noticeably lower. In addition, SO staining is not the only parameter which is included in the OARSI score. We also evaluated the cartilage thickness, proteoglycan structure, and Cartilage surface fibrillation index. Our evaluation to determine the OARSI score relies on the qualities of the whole joint, not only the magnified portion. For convenience we have also outlined the region of positive SO stain in the revised figure 9I

  2. eLife assessment

    This fundamental work advances our understanding of the role of Cbfβ in maintaining articular cartilage homeostasis and the underlying mechanisms. The evidence supporting the conclusion is mostly convincing, although including additional experiments and discussions would have strengthened the study. This paper is of potential interest to skeletal biologists and orthopaedic surgeons who study the pathogenesis and the therapeutics of osteoarthritis.

  3. Reviewer #1 (Public Review):

    Osteoarthritis (OA) is associated with painful, chronic inflammation that often leads to severe joint pain and joint stiffness for people over the age of 55. There is no effective therapeutic drug in the treatment of osteoarthritis. The authors found that mice without Cbfβ in their chondrocytes develop spontaneous OA. Authors uncovered that the deficiency of Cbfβ caused increased canonical Wnt signaling and inflammatory response, and decreased Hippo/YAP signaling and TGF-β signaling in articular cartilage. Authors showed that ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfβ overexpression promoted Yap expression, diminished active β-catenin expression in OA lesions. The authors demonstrated that AAV-mediated Cbfβ overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. The results from the study demonstrated Cbfβ maintains articular cartilage homeostasis through inhibiting Wnt/β-catenin signaling and increasing Hippo/Yap, and TGFβ signaling. Importantly, the authors proved that local Cbfβ overexpression could be an effective strategy for treatment of OA. The data shown in the study demonstrated that the findings are novel and very significant, and the authors' claims and conclusions are justified by their data. The paper is generally excellent with an interesting scientific premise and strong scientific rigor. The findings in this manuscript are novel, the manuscript is clearly written, and the findings will make a significant impact in the field.

  4. Reviewer #2 (Public Review):

    In this manuscript, Chen et al. reported that the core binding factor beta (Cbfβ), a heterodimeric subunit of the RUNX family transcription factors (TFs), is crucial in maintaining cartilage homeostasis and counteracting traumatic OA pathology. Using mouse models in which Cbfβ is conditionally inactivated in the Col2a1+ and Acan+ cells, the authors claimed that Cbfβ ablation led to articular cartilage (AC) degeneration, which is associated with aberrant cartilage gene expression and chondrocyte signaling, particularly the elevated Wnt/Catenin and the decreased Hippo/YAP and TGFβ signaling. The authors further showed that Cbfβ transcripts are decreased in human OA cartilage, and sustaining Cbfβ expression in mouse knee joints mitigated the severity of surgery-evoked OA.

    On the whole, the work reported is interesting and exciting. Genetic and biochemical data support key statements. Both in vivo and in vitro experiments were well designed with proper controls; semiquantitative data were digitalized and processed for statistical significance. Furthermore, new findings were adequately discussed in contrast to the current available knowledge. However, the conceptual novelty of this study is slightly compromised by recent publications showing that Cbfβ reduction is associated with OA (Che et al. 2023; Li et al. 2021). Also, the authors claimed that multiple signaling pathways were affected by Cbfβ ablation in cartilage cells; many of them, however, are indirect effects given the nature of Cbfβ as a TF. The authors also showed that pSMAD2/3 and active βCatenin decreased and increased upon Cbfβ depletion in the mouse AC cartilage. However, how the deficiency of Cbfβ, a widely expressed TF, affected the posttranslational modification of SMAD2/3 and βCatenin is unclear and needs further discussion. Overall, Cbfβ's role in cartilage and OA pathology is an emerging area of study; the authors provided a set of genetic evidences showing that Cbfβ is indispensable for cartilage homeostasis.

  5. Reviewer #3 (Public Review):

    The authors comprehensively demonstrated the Cbfβ gene, which is involved in articular cartilage homeostasis, can promote articular cartilage regeneration and repair in osteoarthritis (OA) through regulating Hippo/YAP signaling TGF-β signaling, and canonical Wnt signaling. First, the authors demonstrated the deletion of Cbfβ can induce the OA phenotypes including decreased articular cartilage and osteoblasts, and increased osteoclasts and subchondral bone hyperplasia, and induce the early onset of OA. Additionally, the authors showed that the deficiency of Cbfβ in cartilage can increase canonical Wnt signaling and decrease TGF-β and Hippo signaling. Finally, the authors demonstrated that the overexpression of Cbfβ can inhibit Wnt signaling and enhance Hippo/YAP signaling in knee joints articular cartilage of ACLT-induced OA mice and protect against ACLT-induced OA. The manuscript is overall well-constructed, and the authors provided evidence to support their findings.

    In Fig. 7I, it could be better to show the statistical analysis between normal and AAV-mediated Cbfβ ACLT mice groups.

    In Fig. 9H-K, in the quantification analysis, the OARSI score in the DMM+AAV-YFP group is higher than in the sham group significantly. However, the SO staining results appear to show no significant difference between the DMM+AAV-luc-YFP group (Fig. 9I) and the sham group (Fig. 9H).