1
|
Komori T. Regulation of Skeletal Development and Maintenance by Runx2 and Sp7. Int J Mol Sci 2024; 25:10102. [PMID: 39337587 PMCID: PMC11432631 DOI: 10.3390/ijms251810102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Runx2 (runt related transcription factor 2) and Sp7 (Sp7 transcription factor 7) are crucial transcription factors for bone development. The cotranscription factor Cbfb (core binding factor beta), which enhances the DNA-binding capacity of Runx2 and stabilizes the Runx2 protein, is necessary for bone development. Runx2 is essential for chondrocyte maturation, and Sp7 is partly involved. Runx2 induces the commitment of multipotent mesenchymal cells to osteoblast lineage cells and enhances the proliferation of osteoprogenitors. Reciprocal regulation between Runx2 and the Hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathways and Dlx5 (distal-less homeobox 5) plays an important role in these processes. The induction of Fgfr2 (Fgf receptor 2) and Fgfr3 expression by Runx2 is important for the proliferation of osteoblast lineage cells. Runx2 induces Sp7 expression, and Runx2+ osteoprogenitors become Runx2+Sp7+ preosteoblasts. Sp7 induces the differentiation of preosteoblasts into osteoblasts without enhancing their proliferation. In osteoblasts, Runx2 is required for bone formation by inducing the expression of major bone matrix protein genes, including Col1a1 (collagen type I alpha 1), Col1a2, Spp1 (secreted phosphoprotein 1), Ibsp (integrin binding sialoprotein), and Bglap (bone gamma carboxyglutamate protein)/Bglap2. Bglap/Bglap2 (osteocalcin) regulates the alignment of apatite crystals parallel to collagen fibrils but does not function as a hormone that regulates glucose metabolism, testosterone synthesis, and muscle mass. Sp7 is also involved in Co1a1 expression and regulates osteoblast/osteocyte process formation, which is necessary for the survival of osteocytes and the prevention of cortical porosity. SP7 mutations cause osteogenesis imperfecta in rare cases. Runx2 is an important pathogenic factor, while Runx1, Runx3, and Cbfb are protective factors in osteoarthritis development.
Collapse
Affiliation(s)
- Toshihisa Komori
- Department of Molecular Tumor Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| |
Collapse
|
2
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
3
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and Tgfβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. eLife 2024; 13:e95640. [PMID: 38805545 PMCID: PMC11132684 DOI: 10.7554/elife.95640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
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 CBFB (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 Cbfb in tamoxifen-induced Cbfbf/f;Col2a1-CreERT 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 Tgfβ 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 Cbfb overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfb 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 Cbfb 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 Cbfb overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
4
|
Zhang Y, Chen H, Wu J, McVicar A, Chen Y, Su J, Li YP, Chen W. Deficiency of Cbfβ in articular cartilage leads to osteoarthritis-like phenotype through Hippo/Yap, TGFβ, and Wnt/β-catenin signaling pathways. Int J Biol Sci 2024; 20:1965-1977. [PMID: 38617544 PMCID: PMC11008268 DOI: 10.7150/ijbs.90250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 01/05/2024] [Indexed: 04/16/2024] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disorder, causing physical impairments among the elderly. Core binding factor subunit β (Cbfβ) has a critical role in bone homeostasis and cartilage development. However, the function and mechanism of Cbfβ in articular cartilage and OA remains unclear. We found that Cbfβf/fAggrecan-CreERT mice with Cbfβ-deficiency in articular cartilage developed a spontaneous osteoarthritis-like phenotype with articular cartilage degradation. Immunofluorescence staining showed that Cbfβf/fAggrecan-CreERT mice exhibited a significant increase in the expression of articular cartilage degradation markers and inflammatory markers in the knee joints. RNA-sequencing analysis demonstrated that Cbfβ orchestrated Hippo/Yap, TGFβ/Smad, and Wnt/β-catenin signaling pathways in articular cartilage, and Cbfβ deficiency resulted in the abnormal expression of downstream genes involved in maintaining articular cartilage homeostasis. Immunofluorescence staining results showed Cbfβ deficiency significantly increased active β-catenin and TCF4 expression while reducing Yap, TGFβ1, and p-Smad 2/3 expression. Western blot and qPCR validated gene expression changes in hip articular cartilage of Cbfβ-deficient mice. Our results demonstrate that deficiency of Cbfβ in articular cartilage leads to an OA-like phenotype via affecting Hippo/Yap, TGFβ, and Wnt/β-catenin signaling pathways, disrupting articular cartilage homeostasis and leading to the pathological process of OA in mice. Our results indicate that targeting Cbfβ may be a potential therapeutic target for the design of novel and effective treatments for OA.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics and Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Shaanxi, Xi'an 710049, P.R. China
| | - Huiwen Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jinjin Wu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, P.R. China
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, 70112, USA
| | - Wei Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, 70112, USA
| |
Collapse
|
5
|
潘 媛, 顾 航, 肖 涵, 赵 笠, 汤 祎, 葛 雯. [Ubiquitin-specific protease 42 regulates osteogenic differentiation of human adipose-derived stem cells]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2024; 56:9-16. [PMID: 38318890 PMCID: PMC10845180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVE To explore the effect of ubiquitin-specific protease 42 (USP42) on osteogenic differentiation of human adipose-derived stem cells (hASCs) in vivo and in vitro. METHODS A combination of experiments was carried out with genetic depletion of USP42 using a lentiviral strategy. Alkaline phosphatase (ALP) staining and quantification, alizarin red S (ARS) staining and quantification were used to determine the osteogenic differentiation ability of hASCs under osteogenic induction between the experimental group (knockdown group and overexpression group) and the control group. Quantitative reverse transcription PCR (qRT-PCR) was used to detect the expression levels of osteogenesis related genes in the experimental group and control group, and Western blotting was used to detect the expression levels of osteogenesis related proteins in the experimental group and control group. Nude mice ectopic implantation experiment was used to evaluate the effect of USP42 on the osteogenic differentiation of hASCs in vivo. RESULTS The mRNA and protein expressions of USP42 in knockdown group were significantly lower than those in control group, and those in overexpression group were significantly higher than those in control group. After 7 days of osteogenic induction, the ALP activity in the knockdown group was significantly higher than that in the control group, and ALP activity in overexpression group was significantly lower than that in control group. After 14 days of osteogenic induction, ARS staining was significantly deeper in the knockdown group than in the control group, and significantly lighter in overexpression group than in the control group. The results of qRT-PCR showed that the mRNA expression levels of ALP, osterix (OSX) and collagen type Ⅰ (COLⅠ) in the knockdown group were significantly higher than those in the control group after 14 days of osteogenic induction, and those in overexpression group were significantly lower than those in control group. The results of Western blotting showed that the expression levels of runt-related transcription factor 2 (RUNX2), OSX and COLⅠ in the knockout group were significantly higher than those in the control group at 14 days after osteogenic induction, while the expression levels of RUNX2, OSX and COLⅠ in the overexpression group were significantly lower than those in the control group. Hematoxylin-eosin staining of subcutaneous grafts in nude mice showed that the percentage of osteoid area in the knockdown group was significantly higher than that in the control group. CONCLUSION Knockdown of USP42 can significantly promote the osteogenic differentiation of hASCs in vitro and in vivo, and overexpression of USP42 significantly inhibits in vivo osteogenic differentiation of hASCs, and USP42 can provide a potential therapeutic target for bone tissue engineering.
Collapse
Affiliation(s)
- 媛 潘
- 北京大学口腔医学院·口腔医院综合二科,国家口腔医学中心,国家口腔疾病临床医学研究中心,口腔生物材料和数字诊疗装备国家工程研究中心,北京 100081Department of General Dentistry Ⅱ, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomate-rials and Digital Medical Devices, Beijing 100081, China
| | - 航 顾
- 北京大学口腔医学院·口腔医院综合二科,国家口腔医学中心,国家口腔疾病临床医学研究中心,口腔生物材料和数字诊疗装备国家工程研究中心,北京 100081Department of General Dentistry Ⅱ, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomate-rials and Digital Medical Devices, Beijing 100081, China
| | - 涵 肖
- 北京大学口腔医学院·口腔医院综合二科,国家口腔医学中心,国家口腔疾病临床医学研究中心,口腔生物材料和数字诊疗装备国家工程研究中心,北京 100081Department of General Dentistry Ⅱ, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomate-rials and Digital Medical Devices, Beijing 100081, China
| | - 笠君 赵
- 北京大学口腔医学院·口腔医院综合二科,国家口腔医学中心,国家口腔疾病临床医学研究中心,口腔生物材料和数字诊疗装备国家工程研究中心,北京 100081Department of General Dentistry Ⅱ, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomate-rials and Digital Medical Devices, Beijing 100081, China
| | - 祎熳 汤
- 北京大学口腔医学院·口腔医院第四门诊部,北京 100025Fourth Clinical Division, Peking University School and Hospital of Stomatology, Beijing 100025, China
| | - 雯姝 葛
- 北京大学口腔医学院·口腔医院综合二科,国家口腔医学中心,国家口腔疾病临床医学研究中心,口腔生物材料和数字诊疗装备国家工程研究中心,北京 100081Department of General Dentistry Ⅱ, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomate-rials and Digital Medical Devices, Beijing 100081, China
| |
Collapse
|
6
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and TGFβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575763. [PMID: 38293189 PMCID: PMC10827176 DOI: 10.1101/2024.01.15.575763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
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 Cbfβf/fCol2α1-CreERT 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.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
7
|
Ma Y, Wang S, Wang H, Chen X, Shuai Y, Wang H, Mao Y, He F. Mesenchymal stem cells and dental implant osseointegration during aging: from mechanisms to therapy. Stem Cell Res Ther 2023; 14:382. [PMID: 38124153 PMCID: PMC10734190 DOI: 10.1186/s13287-023-03611-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023] Open
Abstract
Dental implants are widely used to replace missing teeth, providing patients with unparalleled levels of effectiveness, convenience, and affordability. The biological basis for the clinical success of dental implants is osseointegration. Bone aging is a high-risk factor for the reduced osseointegration and survival rates of dental implants. In aged individuals, mesenchymal stem cells (MSCs) in the bone marrow show imbalanced differentiation with a reduction in osteogenesis and an increase in adipogenesis. This leads to impaired osseointegration and implant failure. This review focuses on the molecular mechanisms underlying the dysfunctional differentiation of aged MSCs, which primarily include autophagy, transcription factors, extracellular vesicle secretion, signaling pathways, epigenetic modifications, microRNAs, and oxidative stress. Furthermore, this review addresses the pathological changes in MSCs that affect osseointegration and discusses potential therapeutic interventions to enhance osseointegration by manipulating the mechanisms underlying MSC aging.
Collapse
Affiliation(s)
- Yang Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Siyuan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Hui Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xiaoyu Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Yi Shuai
- Nanjing Jinling Hospital: East Region Military Command General Hospital, Nanjing, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Yingjie Mao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| | - Fuming He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
8
|
Jo S, Lee SH, Jeon C, Jo HR, Ko E, Whangbo M, Kim TJ, Park YS, Kim TH. Elevated BMPR2 expression amplifies osteoblast differentiation in ankylosing spondylitis. JOURNAL OF RHEUMATIC DISEASES 2023; 30:243-250. [PMID: 37736586 PMCID: PMC10509643 DOI: 10.4078/jrd.2023.0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/31/2023] [Accepted: 06/21/2023] [Indexed: 09/23/2023]
Abstract
Objective Bone morphogenetic protein receptor type 2 (BMPR2) has been associated with radiographic changes in ankylosing spondylitis (AS), but further characterization of the cellular signaling pathway in osteoprogenitor (OP) is not clearly understood. The aim of this study was to investigate the expression of BMPR2 and bone morphogenetic protein 2 (BMP2)-mediated responsibility in AS. Methods We collected 10 healthy control (HC) and 14 AS-OPs derived from facet joints. Subsequently, we then conducted RNA sequencing with two samples per group and selected BMP-related genes. Facet joint tissues and derived primary OPs were evaluated by validation of selected RNA sequencing data, immunohistochemistry, and comparison of osteogenic differentiation potential. Results Based on RNA-sequencing analysis, we found that BMPR2 expression is higher in AS-OPs compared to in HC-OPs. We also validated the increased BMPR2 expression in facet joint tissues with AS and its derived OPs in messenger RNA and protein levels. Additionally, primary AS-OPs showed much greater response to osteogenic differentiation induced by BMP2 and a higher capacity for smad1/5/8-induced RUNX2 expression compared to HCs. Conclusion The expression of BMPR2 was found to be significantly increased in facet joint tissues of patients with AS. These findings suggest that BMPR2 may play a role in the BMP2-mediated progression of AS.
Collapse
Affiliation(s)
- Sungsin Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
| | - Seung Hoon Lee
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
| | - Chanhyeok Jeon
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
- Department of Translational Medicine Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Hye-Ryeong Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
| | - Eunae Ko
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
- Department of Translational Medicine Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Min Whangbo
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
- Department of Translational Medicine Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Tae-Jong Kim
- Department of Rheumatology, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Ye-Soo Park
- Department of Orthopedic Surgery, Hanyang University Guri Hospital, Hanyang University College of Medicine, Guri, Korea
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, Korea
- Department of Translational Medicine Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, Korea
| |
Collapse
|
9
|
Che X, Jin X, Park NR, Kim HJ, Kyung HS, Kim HJ, Lian JB, Stein JL, Stein GS, Choi JY. Cbfβ Is a Novel Modulator against Osteoarthritis by Maintaining Articular Cartilage Homeostasis through TGF-β Signaling. Cells 2023; 12:cells12071064. [PMID: 37048137 PMCID: PMC10093452 DOI: 10.3390/cells12071064] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
TGF-β signaling is a vital regulator for maintaining articular cartilage homeostasis. Runx transcription factors, downstream targets of TGF-β signaling, have been studied in the context of osteoarthritis (OA). Although Runx partner core binding factor β (Cbfβ) is known to play a pivotal role in chondrocyte and osteoblast differentiation, the role of Cbfβ in maintaining articular cartilage integrity remains obscure. This study investigated Cbfβ as a novel anabolic modulator of TGF-β signaling and determined its role in articular cartilage homeostasis. Cbfβ significantly decreased in aged mouse articular cartilage and human OA cartilage. Articular chondrocyte-specific Cbfb-deficient mice (Cbfb△ac/△ac) exhibited early cartilage degeneration at 20 weeks of age and developed OA at 12 months. Cbfb△ac/△ac mice showed enhanced OA progression under the surgically induced OA model in mice. Mechanistically, forced expression of Cbfβ rescued Type II collagen (Col2α1) and Runx1 expression in Cbfβ-deficient chondrocytes. TGF-β1-mediated Col2α1 expression failed despite the p-Smad3 activation under TGF-β1 treatment in Cbfβ-deficient chondrocytes. Cbfβ protected Runx1 from proteasomal degradation through Cbfβ/Runx1 complex formation. These results indicate that Cbfβ is a novel anabolic regulator for cartilage homeostasis, suggesting that Cbfβ could protect OA development by maintaining the integrity of the TGF-β signaling pathway in articular cartilage.
Collapse
|
10
|
Cho E, Che X, Ang MJ, Cheon S, Lee J, Kim KS, Lee CH, Lee SY, Yang HY, Moon C, Park C, Choi JY, Lee TH. Peroxiredoxin 5 regulates osteogenic differentiation through interaction with hnRNPK during bone regeneration. eLife 2023; 12:80122. [PMID: 36735291 PMCID: PMC9897727 DOI: 10.7554/elife.80122] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 01/18/2023] [Indexed: 02/04/2023] Open
Abstract
Peroxiredoxin 5 (Prdx5) is involved in pathophysiological regulation via the stress-induced cellular response. However, its function in the bone remains largely unknown. Here, we show that Prdx5 is involved in osteoclast and osteoblast differentiation, resulting in osteoporotic phenotypes in Prdx5 knockout (Prdx5Ko) male mice. To investigate the function of Prdx5 in the bone, osteoblasts were analyzed through immunoprecipitation (IP) and liquid chromatography combined with tandem mass spectrometry (LC-MS/MS) methods, while osteoclasts were analyzed through RNA-sequencing. Heterogeneous nuclear ribonucleoprotein K (hnRNPK) was identified as a potential binding partner of Prdx5 during osteoblast differentiation in vitro. Prdx5 acts as a negative regulator of hnRNPK-mediated osteocalcin (Bglap) expression. In addition, transcriptomic analysis revealed that in vitro differentiated osteoclasts from the bone marrow-derived macrophages of Prdx5Ko mice showed enhanced expression of several osteoclast-related genes. These findings indicate that Prdx5 might contribute to the maintenance of bone homeostasis by regulating osteoblast differentiation. This study proposes a new function of Prdx5 in bone remodeling that may be used in developing therapeutic strategies for bone diseases.
Collapse
Affiliation(s)
- Eunjin Cho
- Department of Oral Biochemistry, Korea Mouse Phenotype Center (KMPC), Dental Science Research Institute, School of Dentistry, Chonnam National UniversityGwangjuRepublic of Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, Skeletal Diseases Analysis Center, Korea Mouse Phenotyping Center (KMPC), School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Mary Jasmin Ang
- Department of Basic Veterinary Sciences, College of Veterinary Medicine, University of the Philippines Los BañosLos BañosPhilippines
| | - Seongmin Cheon
- School of Biological Sciences and Technology, Chonnam National UniversityGwangjuRepublic of Korea,Proteomics Core Facility, Biomedical Research Institute, Seoul National University HospitalSeoulRepublic of Korea
| | - Jinkyung Lee
- Department of Oral Biochemistry, Korea Mouse Phenotype Center (KMPC), Dental Science Research Institute, School of Dentistry, Chonnam National UniversityGwangjuRepublic of Korea
| | - Kwang Soo Kim
- Department of Microbiology, Department of Molecular Medicine (BK21plus), Chonnam National University Medical SchoolGwangjuRepublic of Korea
| | - Chang Hoon Lee
- Therapeutic & Biotechnology Division, Drug Discovery Platform Research Center, Research Institute of Chemical Technology (KRICT)DaejeonRepublic of Korea
| | - Sang-Yeop Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science InstituteOchangRepublic of Korea
| | - Hee-Young Yang
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation FoundationDaeguRepublic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National UniversityGwangjuRepublic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National UniversityGwangjuRepublic of Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, BK21 Plus KNU Biomedical Convergence Program, Skeletal Diseases Analysis Center, Korea Mouse Phenotyping Center (KMPC), School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Tae-Hoon Lee
- Department of Oral Biochemistry, Korea Mouse Phenotype Center (KMPC), Dental Science Research Institute, School of Dentistry, Chonnam National UniversityGwangjuRepublic of Korea
| |
Collapse
|
11
|
Jiang Q, Qin X, Nagano K, Komori H, Matsuo Y, Taniuchi I, Ito K, Komori T. Different Requirements of CBFB and RUNX2 in Skeletal Development among Calvaria, Limbs, Vertebrae and Ribs. Int J Mol Sci 2022; 23:13299. [PMID: 36362086 PMCID: PMC9657020 DOI: 10.3390/ijms232113299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 10/01/2023] Open
Abstract
RUNX proteins, such as RUNX2, regulate the proliferation and differentiation of chondrocytes and osteoblasts. Haploinsufficiency of RUNX2 causes cleidocranial dysplasia, but a detailed analysis of Runx2+/- mice has not been reported. Furthermore, CBFB is required for the stability and DNA binding of RUNX family proteins. CBFB has two isoforms, and CBFB2 plays a major role in skeletal development. The calvaria, femurs, vertebrae and ribs in Cbfb2-/- mice were analyzed after birth, and compared with those in Runx2+/- mice. Calvarial development was impaired in Runx2+/- mice but mildly delayed in Cbfb2-/- mice. In femurs, the cortical bone but not trabecular bone was reduced in Cbfb2-/- mice, whereas both the trabecular and cortical bone were reduced in Runx2+/- mice. The trabecular bone in vertebrae increased in Cbfb2-/- mice but not in Runx2+/- mice. Rib development was impaired in Cbfb2-/- mice but not in Runx2+/- mice. These differences were likely caused by differences in the indispensability of CBFB and RUNX2, the balance of bone formation and resorption, or the number and maturation stage of osteoblasts. Thus, different amounts of CBFB and RUNX2 were required among the bone tissues for proper bone development and maintenance.
Collapse
Affiliation(s)
- Qing Jiang
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Institute of Orthopaedics, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Xin Qin
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Institute of Orthopaedics, Suzhou Medical College, Soochow University, Suzhou 215006, China
| | - Kenichi Nagano
- Department of Oral Pathology and Bone Metabolism, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hisato Komori
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Yuki Matsuo
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama 230-0045, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Toshihisa Komori
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
12
|
Wu Q, Li B, Li Y, Liu F, Yang L, Ma Y, Zhang Y, Xu D, Li Y. Effects of PAMK on lncRNA, miRNA, and mRNA expression profiles of thymic epithelial cells. Funct Integr Genomics 2022; 22:849-863. [PMID: 35505120 DOI: 10.1007/s10142-022-00863-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 11/04/2022]
Abstract
Polysaccharides from Atractylodes macrocephala Koidz (PAMK) can promote the proliferation of thymocytes and improve the body's immunity. However, the effect of PAMK on thymic epithelial cells has not been reported. Studies have shown that miRNAs and lncRNAs are key factors in regulating cell proliferation. In this study, we found that PAMK could promote the proliferation of mouse medullary thymic epithelial cell line 1 (MTEC1) cells through CCK-8 and EdU experiments. To further explore its mechanism, we detected the effect of PAMK on the expression profiles of lncRNAs, miRNAs, and mRNAs in MTEC1 cells. The results showed that PAMK significantly affected the expression of 225 lncRNAs, 29 miRNAs, and 800 mRNAs. Functional analysis showed that these differentially expressed genes were significantly enriched in cell cycle, cell division, NF-kappaB signaling, apoptotic process, and MAPK signaling pathway. Finally, we used Cytoscape to visualize lncRNA-miRNA-mRNA(14 lncRNAs, 17 miRNAs, 171 mRNAs) networks based on ceRNA theory. These results suggest that lncRNAs and miRNAs may be involved in the effect of PAMK on the proliferation of MTEC1 cells, providing a new research direction for exploring the molecular mechanism of PAMK promoting the proliferation of thymic epithelial cells.
Collapse
Affiliation(s)
- Qingru Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Bingxin Li
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Fenfen Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Lin Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yongjiang Ma
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yuan Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Danning Xu
- Guangdong Province Key Laboratory of Waterfowl Healthy Breeding, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China.
| | - Yugu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
13
|
Kim HJ, Lee DK, Jin X, Che X, Ryu SH, Choi JY. Phospholipase D2 controls bone homeostasis by modulating M-CSF-dependent osteoclastic cell migration and microtubule stability. EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:1146-1155. [PMID: 35945449 PMCID: PMC9440116 DOI: 10.1038/s12276-022-00820-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022]
Abstract
Phospholipase D2 (PLD2), a signaling protein, plays a central role in cellular communication and various biological processes. Here, we show that PLD2 contributes to bone homeostasis by regulating bone resorption through osteoclastic cell migration and microtubule-dependent cytoskeletal organization. Pld2-deficient mice exhibited a low bone mass attributed to increased osteoclast function without altered osteoblast activity. While Pld2 deficiency did not affect osteoclast differentiation, its absence promoted the migration of osteoclast lineage cells through a mechanism involving M-CSF-induced activation of the PI3K–Akt–GSK3β signaling pathway. The absence of Pld2 also boosted osteoclast spreading and actin ring formation, resulting in elevated bone resorption. Furthermore, Pld2 deletion increased microtubule acetylation and stability, which were later restored by treatment with a specific inhibitor of Akt, an essential molecule for microtubule stabilization and osteoclast bone resorption activity. Interestingly, PLD2 interacted with the M-CSF receptor (c-Fms) and PI3K, and the association between PLD2 and c-Fms was reduced in response to M-CSF. Altogether, our findings indicate that PLD2 regulates bone homeostasis by modulating osteoclastic cell migration and microtubule stability via the M-CSF-dependent PI3K–Akt–GSK3β axis. A signaling protein that regulates bone resorption may prove a useful target in treating skeletal conditions such as osteoporosis and rheumatoid arthritis. Bone is synthesized by cells called osteoblasts, while osteoclasts trigger bone resorption, keeping the skeleton healthy. Imbalances in this recycling process are common in bone disorders. Je-Young Choi and Hyun-Ju Kim at Kyungpook National University in Daegu, South Korea, and co-workers demonstrated that phospholipase D2 (PLD2), a membrane protein, directly regulates bone resorption in mice. Mice without the Pld2 gene had increased osteoclast activity, resulting in low bone mass. The absence of PLD2 promotes the migration of osteoclasts via a particular signaling pathway. This increased the organization of microtubules, polymers that help form the cytoskeleton. The results suggest that regulating PLD2 activity could form the basis of a future treatment method.
Collapse
Affiliation(s)
- Hyun-Ju Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| | - Dong-Kyo Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Xian Jin
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sung Ho Ryu
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, BK21 Plus KNU Biomedical Convergence Program, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
14
|
Whole Aspect of Runx2 Functions in Skeletal Development. Int J Mol Sci 2022; 23:ijms23105776. [PMID: 35628587 PMCID: PMC9144571 DOI: 10.3390/ijms23105776] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Runt-related transcription factor 2 (Runx2) is a fundamental transcription factor for bone development. In endochondral ossification, Runx2 induces chondrocyte maturation, enhances chondrocyte proliferation through Indian hedgehog (Ihh) induction, and induces the expression of vascular endothelial growth factor A (Vegfa), secreted phosphoprotein 1 (Spp1), integrin-binding sialoprotein (Ibsp), and matrix metallopeptidase 13 (Mmp13) in the terminal hypertrophic chondrocytes. Runx2 inhibits the apoptosis of the terminal hypertrophic chondrocytes and induces their transdifferentiation into osteoblasts and osteoblast progenitors. The transdifferentiation is required for trabecular bone formation during embryonic and newborn stages but is dispensable for acquiring normal bone mass in young and adult mice. Runx2 enhances the proliferation of osteoblast progenitors and induces their commitment to osteoblast lineage cells through the direct regulation of the expressions of a hedgehog, fibroblast growth factor (Fgf), Wnt, and parathyroid hormone-like hormone (Pthlh) signaling pathway genes and distal-less homeobox 5 (Dlx5), which all regulate Runx2 expression and/or protein activity. Runx2, Sp7, and Wnt signaling further induce osteoblast differentiation. In immature osteoblasts, Runx2 regulates the expression of bone matrix protein genes, including Col1a1, Col1a2, Spp1, Ibsp, and bone gamma carboxyglutamate protein (Bglap)/Bglap2, and induces osteoblast maturation. Osteocalcin (Bglap/Bglap2) is required for the alignment of apatite crystals parallel to the collagen fibers; however, it does not physiologically work as a hormone that regulates glucose metabolism, testosterone synthesis, or muscle mass. Thus, Runx2 exerts multiple functions essential for skeletal development.
Collapse
|
15
|
Osteoblast-derived vesicles induce a switch from bone-formation to bone-resorption in vivo. Nat Commun 2022; 13:1066. [PMID: 35210428 PMCID: PMC8873258 DOI: 10.1038/s41467-022-28673-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
Bone metabolism is regulated by the cooperative activity between bone-forming osteoblasts and bone-resorbing osteoclasts. However, the mechanisms mediating the switch between the osteoblastic and osteoclastic phases have not been fully elucidated. Here, we identify a specific subset of mature osteoblast-derived extracellular vesicles that inhibit bone formation and enhance osteoclastogenesis. Intravital imaging reveals that mature osteoblasts secrete and capture extracellular vesicles, referred to as small osteoblast vesicles (SOVs). Co-culture experiments demonstrate that SOVs suppress osteoblast differentiation and enhance the expression of receptor activator of NF-κB ligand, thereby inducing osteoclast differentiation. We also elucidate that the SOV-enriched microRNA miR-143 inhibits Runt-related transcription factor 2, a master regulator of osteoblastogenesis, by targeting the mRNA expression of its dimerization partner, core-binding factor β. In summary, we identify SOVs as a mode of cell-to-cell communication, controlling the dynamic transition from bone-forming to bone-resorbing phases in vivo.
Collapse
|
16
|
Che X, Park NR, Jin X, Jung YK, Han MS, Park CY, Chun JS, Kim SG, Jin J, Kim HJ, Lian JB, Stein JL, Stein GS, Choi JY. Hypoxia-inducible factor 2α is a novel inhibitor of chondrocyte maturation. J Cell Physiol 2021; 236:6963-6973. [PMID: 33748969 PMCID: PMC8662706 DOI: 10.1002/jcp.30356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022]
Abstract
Hypoxic environment is essential for chondrocyte maturation and longitudinal bone growth. Although hypoxia-inducible factor 1 alpha (Hif-1α) has been known as a key player for chondrocyte survival and function, the function of Hif-2α in cartilage is mechanistically and clinically relevant but remains unknown. Here we demonstrated that Hif-2α was a novel inhibitor of chondrocyte maturation through downregulation of Runx2 stability. Mechanistically, Hif-2α binding to Runx2 inhibited chondrocyte maturation by Runx2 degradation through disrupting Runx2/Cbfβ complex formation. The Hif-2α-mediated-Runx2 degradation could be rescued by Cbfβ transfection due to the increase of Runx2/Cbfβ complex formation. Consistently, mesenchymal cells derived from Hif-2α heterozygous mice were more rapidly differentiated into hypertrophic chondrocytes than those of wild-type mice in a micromass culture system. Collectively, these findings demonstrate that Hif-2α is a novel inhibitor for chondrocyte maturation by disrupting Runx2/Cbfβ complex formation and consequential regulatory activity.
Collapse
Affiliation(s)
- Xiangguo Che
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Na-Rae Park
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Xian Jin
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Youn-Kwan Jung
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Min-Su Han
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Clara Yongjoo Park
- Department of Food and Nutrition, Human Ecology Research Institute, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jang-Soo Chun
- Cell Dynamics Research Center, School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Seong-Gon Kim
- Department of Oral and Maxillofacial Surgery, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, Republic of Korea
| | - Jingchun Jin
- Department of Immunology of Yanbian University Hospital, 133000, Yanji, Jilin Province, China
| | - Hyun-Ju Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| | - Jane B. Lian
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Brulington, VT 05405, U.S.A
| | - Janet L. Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Brulington, VT 05405, U.S.A
| | - Gary S. Stein
- Department of Biochemistry and University of Vermont Cancer Center, University of Vermont College of Medicine, Brulington, VT 05405, U.S.A
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, Korea Mouse Phenotyping Center, School of Medicine, Kyungpook National University, Daegu 41944 Korea
| |
Collapse
|
17
|
Li F, Wu H, Zou G, Cang D, Shen Y. Circular RNA_0062582 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells via regulation of microRNA-145/CBFB axis. Bioengineered 2021; 12:1952-1963. [PMID: 34027799 PMCID: PMC8806255 DOI: 10.1080/21655979.2021.1921553] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Osteoporosis poses a threat to human health worldwide. To date, there have been few studies regarding targeted treatment of osteoporosis. We aimed to identify the possible molecular mechanism of circular RNA (circ)_0062582 in osteogenic differentiation, and the interactions among circ_0062582, microRNA-145 (miR-145) and core-binding factor subunit β (CBFB). The proliferation of human bone marrow mesenchymal stem cells (hBMSCs) was tested with a cell counting kit-8 assay. Circ_0062582, miR-145 and CBFB were overexpressed by transient transfection. Dual-luciferase reporter assay system was used to analyze the combination among circ_0062582, miR-145 and CBFB. Additionally, the levels of circ_0062582, miR-145, CBFB, osterix (OSX), osteocalcin (OCN) and collagen type 1 (COL1) were detected by means of RT-qPCR or western blot analysis. Alkaline phosphatase and Alizarin red stainings were performed to analyze the degree of osteogenic differentiation under the control of circ_0062582, miR-145 and CBFB. The results demonstrated that circ_0062582 level was notably elvated during osteogenic differentiation of hBMSCs. Circ_0062582 overexpression significantly promoted osteogenic differentiation and upregulated the levels of osteogenic differentiation-related proteins, including OSX, OCN and COL1. In addition, miR-145, which was identified as the target gene of circ_0062582, could specifically target CBFB 3′-UTR regions. Next, these changes caused by the overexpression of circ_0062582 were reversed following the addition of miR-145 mimic. Following overexpression of CBFB, osteogenic differentiation was increased. In summary, these results demonstrated that the role of circ_0062582 in osteoporosis is mediated through regulating the expression level of CBFB via miR-145.
Collapse
Affiliation(s)
- Feng Li
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R.China.,Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, Jiangsu, P.R.China
| | - Hao Wu
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, Jiangsu, P.R.China
| | - Guoyou Zou
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, Jiangsu, P.R.China
| | - Dingwei Cang
- Department of Orthopedics, The First People's Hospital of Yancheng, Yancheng, Jiangsu, P.R.China
| | - Yixin Shen
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, P.R.China
| |
Collapse
|
18
|
隋 昊, 张 陶. [The Role of Histone Demethylase in Osteogenic and Chondrogenic Differentiation of Mesenchymal Stem Cells: A Literature Review]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2021; 52:364-372. [PMID: 34018352 PMCID: PMC10409206 DOI: 10.12182/20210560202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 02/05/2023]
Abstract
The proliferation and multi-directional differentiation potential of mesenchymal stem cells (MSCs) enabled its wide use in the development of new therapies for bone and cartilage repair. Although preliminary work has been done to verify the gene expression profile of MSCs osteogenic and chondrogenic differentiation, it is still unclear what key factors initiate the differentiation of MSCs, resulting in its limited application in bone and cartilage tissue engineering. The epigenetic mechanism mediated by histone demethylases (lysine [K]-specific histone demethylases, KDMs) is the key link in regulating MSCs lineage differentiation. The lysine-specific histone demethylase (LSD) family containing Tower domain and the histone demethylase family containing Jumonji C (JmjC) domain regulate the expression of various osteogenic-related genes, including Runt-related transcription factor 2 ( RUNX2), osterix ( OSX), osteocalcin ( OCN), to mediate MSCs osteogenic differentiation. The KDM2/4/6 subfamilies regulate the chondrogenic differentiation of MSCs through multiple pathways centered on SRY-related high-mobility-group-box gene 9 ( SOX9). In addition, nanotopology, mircoRNAs, etc. regulate the expression of a variety of osteogenic and chondrogenic transcription factors through up- and down-regulation of KDMs. In summary, the role of histone demethylase in the osteogenic and chondrogenic differentiation of mesenchymal stem cells will help us better understand the pathogenesis of bone and cartilage damage diseases, and establish the foundation of future clinical applications for bone and cartilage tissue engineering.
Collapse
Affiliation(s)
- 昊 隋
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - 陶 张
- 口腔疾病研究国家重点实验室 国家口腔疾病临床医学研究中心 四川大学华西口腔医院 (成都 610041)State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
19
|
Xia C, Jiang T, Wang Y, Chen X, Hu Y, Gao Y. The p53/miR-145a Axis Promotes Cellular Senescence and Inhibits Osteogenic Differentiation by Targeting Cbfb in Mesenchymal Stem Cells. Front Endocrinol (Lausanne) 2021; 11:609186. [PMID: 33505358 PMCID: PMC7829338 DOI: 10.3389/fendo.2020.609186] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
The osteogenic differentiation capacity of senescent bone marrow mesenchymal stem cells (MSCs) is reduced. p53 not only regulates cellular senescence but also functions as a negative regulator in bone formation. However, the role of p53 in MSCs senescence and differentiation has not been extensively explored. In the present study, we investigated the molecular mechanism of p53 in MSCs senescence and osteogenic differentiation. We found that p53 was upregulated during cellular senescence and osteogenic differentiation of MSCs respectively induced by H2O2 and BMP9. Similarly, the expression of p53-induced miR-145a was increased significantly. Furthermore, Overexpression of miR-145a in MSCs promoted cellular senescence and inhibited osteogenic differentiation. Then, we identified that p53-induced miR-145a inhibited osteogenic differentiation by targeting core binding factor beta (Cbfb), and the restoration of Cbfb expression rescued the inhibitory effects of miRNA-145a. In summary, our results indicate that p53/miR-145a axis exert its functions both in promoting senescence and inhibiting osteogenesis of MSCs, and the novel p53/miR-145a/Cbfb axis in osteogenic differentiation of MSCs may represent new targets in the treatment of osteoporosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Yanhong Gao
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
20
|
Komori T. Molecular Mechanism of Runx2-Dependent Bone Development. Mol Cells 2020; 43:168-175. [PMID: 31896233 PMCID: PMC7057844 DOI: 10.14348/molcells.2019.0244] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 12/03/2019] [Indexed: 01/09/2023] Open
Abstract
Runx2 is an essential transcription factor for skeletal development. It is expressed in multipotent mesenchymal cells, osteoblast-lineage cells, and chondrocytes. Runx2 plays a major role in chondrocyte maturation, and Runx3 is partly involved. Runx2 regulates chondrocyte proliferation by directly regulating Ihh expression. It also determines whether chondrocytes become those that form transient cartilage or permanent cartilage, and functions in the pathogenesis of osteoarthritis. Runx2 is essential for osteoblast differentiation and is required for the proliferation of osteoprogenitors. Ihh is required for Runx2 expression in osteoprogenitors, and hedgehog signaling and Runx2 induce the differentiation of osteoprogenitors to preosteoblasts in endochondral bone. Runx2 induces Sp7 expression, and Runx2, Sp7, and canonical Wnt signaling are required for the differentiation of preosteoblasts to immature osteoblasts. It also induces the proliferation of osteoprogenitors by directly regulating the expression of Fgfr2 and Fgfr3. Furthermore, Runx2 induces the proliferation of mesenchymal cells and their commitment into osteoblast-lineage cells through the induction of hedgehog (Gli1, Ptch1, Ihh), Fgf (Fgfr2, Fgfr3), Wnt (Tcf7, Wnt10b), and Pthlh (Pth1r) signaling pathway gene expression in calvaria, and more than a half-dosage of Runx2 is required for their expression. This is a major cause of cleidocranial dysplasia, which is caused by heterozygous mutation of RUNX2. Cbfb, which is a co-transcription factor that forms a heterodimer with Runx2, enhances DNA binding of Runx2 and stabilizes Runx2 protein by inhibiting its ubiquitination. Thus, Runx2/Cbfb regulates the proliferation and differentiation of chondrocytes and osteoblast-lineage cells by activating multiple signaling pathways and via their reciprocal regulation.
Collapse
Affiliation(s)
- Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| |
Collapse
|
21
|
Fan W, Yuan L, Li J, Wang Z, Chen J, Guo C, Mo X, Yan Z. Injectable double-crosslinked hydrogels with kartogenin-conjugated polyurethane nano-particles and transforming growth factor β3 for in-situ cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110705. [PMID: 32204019 DOI: 10.1016/j.msec.2020.110705] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/28/2019] [Accepted: 01/28/2020] [Indexed: 01/10/2023]
Abstract
Articular cartilage has a limited ability for self-repair after injury. Implantation of scaffolds functionalized with bioactive molecules that could induce the migration and chondrogenesis of endogenous mesenchymal stem cells (MSCs) provides a convenient alternative for in-situ cartilage regeneration. In this study, we found the synergistic effects of kartogenin (KGN) and transforming growth factor β3 (TGF-β3) on chondrogenesis of MSCs in vitro, indicating that KGN and TGF-β3 are a good match for cartilage regeneration. Furthermore, we confirmed that KGN promoted the chondrogenesis of MSCs through attenuating the degradation of Runx1, which physically interacted with p-Smad3 in nuclei of MSCs. Meanwhile, we designed an injectable double-crosslinked hydrogel with superior mechanical property and longer support for cartilage regeneration by modifying sodium alginate and gelatin. When loaded with KGN conjugated polyurethane nanoparticles (PN-KGN) and TGF-β3, this hydrogel showed biological functions by the release of KGN and TGF-β3, which promoted the MSC migration and cartilage regeneration in one system. In conclusion, the cell-free hydrogel, along with PN-KGN and TGF-β3, provides a promising strategy for cartilage repair by attracting endogenous MSCs and inducing chondrogenesis of recruited cells in a single-step procedure.
Collapse
Affiliation(s)
- Wenshuai Fan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liu Yuan
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China
| | - Jinghuan Li
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jifei Chen
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Changan Guo
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiumei Mo
- Biomaterials and Tissue Engineering Lab, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China; State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, China.
| | - Zuoqin Yan
- Department of Orthopedics, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Šuštić T, Bosdriesz E, van Wageningen S, Wessels LFA, Bernards R. RUNX2/CBFB modulates the response to MEK inhibitors through activation of receptor tyrosine kinases in KRAS-mutant colorectal cancer. Transl Oncol 2019; 13:201-211. [PMID: 31865182 PMCID: PMC6931198 DOI: 10.1016/j.tranon.2019.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 10/10/2019] [Indexed: 10/26/2022] Open
Abstract
Intrinsic and acquired resistances are major hurdles preventing the effective use of MEK inhibitors for treatment of colorectal cancer (CRC). Some 35-45% of colorectal cancers are KRAS-mutant and their treatment remains challenging as these cancers are refractory to MEK inhibitor treatment, because of feedback activation of receptor tyrosine kinases (RTKs). We reported previously that loss of ERN1 sensitizes a subset of KRAS-mutant colon cancer cells to MEK inhibition. Here we show that the loss of RUNX2 or its cofactor CBFB can confer MEK inhibitor resistance in CRC cells. Mechanistically, we find that cells with genetically ablated RUNX2 or CBFB activate multiple RTKs, which coincides with high SHP2 phosphatase activity, a phosphatase that relays signals from the cell membrane to downstream pathways governing growth and proliferation. Moreover, we show that high activity of SHP2 is causal to loss of RUNX2-induced MEK inhibitor resistance, as a small molecule SHP2 inhibitor reinstates sensitivity to MEK inhibitor in RUNX2 knockout cells. Our results reveal an unexpected role for loss of RUNX2/CBFB in regulating RTK activity in colon cancer, resulting in reduced sensitivity to MEK inhibitors.
Collapse
Affiliation(s)
- Tonći Šuštić
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066, CX, the Netherlands
| | - Evert Bosdriesz
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066, CX, the Netherlands
| | - Sake van Wageningen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066, CX, the Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066, CX, the Netherlands
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066, CX, the Netherlands.
| |
Collapse
|
23
|
Feng Y, Liao Y, Zhang J, Shen J, Shao Z, Hornicek F, Duan Z. Transcriptional activation of CBFβ by CDK11 p110 is necessary to promote osteosarcoma cell proliferation. Cell Commun Signal 2019; 17:125. [PMID: 31610798 PMCID: PMC6792216 DOI: 10.1186/s12964-019-0440-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Aberrant expression of cyclin-dependent protein kinases (CDK) is a hallmark of cancer. CDK11 plays a crucial role in cancer cell growth and proliferation. However, the molecular mechanisms of CDK11 and CDK11 transcriptionally regulated genes are largely unknown. METHODS In this study, we performed a global transcriptional analysis using gene array technology to investigate the transcriptional role of CDK11 in osteosarcoma. The promoter luciferase assay, chromatin immunoprecipitation assay, and Gel Shift assay were used to identify direct transcriptional targets of CDK11. Clinical relevance and function of core-binding factor subunit beta (CBFβ) were further accessed in osteosarcoma. RESULTS We identified a transcriptional role of protein-DNA interaction for CDK11p110, but not CDK11p58, in the regulation of CBFβ expression in osteosarcoma cells. The CBFβ promoter luciferase assay, chromatin immunoprecipitation assay, and Gel Shift assay confirmed that CBFβ is a direct transcriptional target of CDK11. High expression of CBFβ is associated with poor outcome in osteosarcoma patients. Expression of CBFβ contributes to the proliferation and metastatic behavior of osteosarcoma cells. CONCLUSIONS These data establish CBFβ as a mediator of CDK11p110 dependent oncogenesis and suggest that targeting the CDK11- CBFβ pathway may be a promising therapeutic strategy for osteosarcoma treatment.
Collapse
Affiliation(s)
- Yong Feng
- Department of Orthopaedic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, 430022 China
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| | - Yunfei Liao
- Department of Orthopaedic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, 430022 China
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| | - Jianming Zhang
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| | - Jacson Shen
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| | - Zengwu Shao
- Department of Orthopaedic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jie Fang Avenue, Wuhan, 430022 China
| | - Francis Hornicek
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Department of Orthopaedic Surgery, Department of Orthopaedic Surgery, David Geffen School of Medicine at UCLA, 615 Charles E. Young Dr. S, Los Angeles, CA 90095 USA
| |
Collapse
|
24
|
Controlling hypoxia-inducible factor-2α is critical for maintaining bone homeostasis in mice. Bone Res 2019; 7:14. [PMID: 31098335 PMCID: PMC6513851 DOI: 10.1038/s41413-019-0054-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Pathological bone loss is caused by an imbalance between bone formation and resorption. The bone microenvironments are hypoxic, and hypoxia-inducible factor (HIF) is known to play notable roles in bone remodeling. However, the relevant functions of HIF-2α are not well understood. Here, we have shown that HIF-2α deficiency in mice enhances bone mass through its effects on the differentiation of osteoblasts and osteoclasts. In vitro analyses revealed that HIF-2α inhibits osteoblast differentiation by targeting Twist2 and stimulates RANKL-induced osteoclastogenesis via regulation of Traf6. In addition, HIF-2α appears to contribute to the crosstalk between osteoblasts and osteoclasts by directly targeting RANKL in osteoprogenitor cells. Experiments performed with osteoblast- and osteoclast-specific conditional knockout mice supported a role of HIF-2α in this crosstalk. HIF-2α deficiency alleviated ovariectomy-induced bone loss in mice, and specific inhibition of HIF-2α with ZINC04179524 significantly blocked RANKL-mediated osteoclastogenesis. Collectively, our results suggest that HIF-2α functions as a catabolic regulator in bone remodeling, which is critical for the maintenance of bone homeostasis.
Collapse
|
25
|
Komori T. Regulation of Proliferation, Differentiation and Functions of Osteoblasts by Runx2. Int J Mol Sci 2019; 20:ijms20071694. [PMID: 30987410 PMCID: PMC6480215 DOI: 10.3390/ijms20071694] [Citation(s) in RCA: 433] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 11/25/2022] Open
Abstract
Runx2 is essential for osteoblast differentiation and chondrocyte maturation. During osteoblast differentiation, Runx2 is weakly expressed in uncommitted mesenchymal cells, and its expression is upregulated in preosteoblasts, reaches the maximal level in immature osteoblasts, and is down-regulated in mature osteoblasts. Runx2 enhances the proliferation of osteoblast progenitors by directly regulating Fgfr2 and Fgfr3. Runx2 enhances the proliferation of suture mesenchymal cells and induces their commitment into osteoblast lineage cells through the direct regulation of hedgehog (Ihh, Gli1, and Ptch1), Fgf (Fgfr2 and Fgfr3), Wnt (Tcf7, Wnt10b, and Wnt1), and Pthlh (Pthr1) signaling pathway genes, and Dlx5. Runx2 heterozygous mutation causes open fontanelle and sutures because more than half of the Runx2 gene dosage is required for the induction of these genes in suture mesenchymal cells. Runx2 regulates the proliferation of osteoblast progenitors and their differentiation into osteoblasts via reciprocal regulation with hedgehog, Fgf, Wnt, and Pthlh signaling molecules, and transcription factors, including Dlx5 and Sp7. Runx2 induces the expression of major bone matrix protein genes, including Col1a1, Spp1, Ibsp, Bglap2, and Fn1, in vitro. However, the functions of Runx2 in differentiated osteoblasts in the expression of these genes in vivo require further investigation.
Collapse
Affiliation(s)
- Toshihisa Komori
- Basic and Translational Research Center for Hard Tissue Disease, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan.
| |
Collapse
|
26
|
Šuštić T, van Wageningen S, Bosdriesz E, Reid RJD, Dittmar J, Lieftink C, Beijersbergen RL, Wessels LFA, Rothstein R, Bernards R. A role for the unfolded protein response stress sensor ERN1 in regulating the response to MEK inhibitors in KRAS mutant colon cancers. Genome Med 2018; 10:90. [PMID: 30482246 PMCID: PMC6258447 DOI: 10.1186/s13073-018-0600-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/13/2018] [Indexed: 12/25/2022] Open
Abstract
Background Mutations in KRAS are frequent in human cancer, yet effective targeted therapeutics for these cancers are still lacking. Attempts to drug the MEK kinases downstream of KRAS have had limited success in clinical trials. Understanding the specific genomic vulnerabilities of KRAS-driven cancers may uncover novel patient-tailored treatment options. Methods We first searched for synthetic lethal (SL) genetic interactions with mutant RAS in yeast with the ultimate aim to identify novel cancer-specific targets for therapy. Our method used selective ploidy ablation, which enables replication of cancer-specific gene expression changes in the yeast gene disruption library. Second, we used a genome-wide CRISPR/Cas9-based genetic screen in KRAS mutant human colon cancer cells to understand the mechanistic connection between the synthetic lethal interaction discovered in yeast and downstream RAS signaling in human cells. Results We identify loss of the endoplasmic reticulum (ER) stress sensor IRE1 as synthetic lethal with activated RAS mutants in yeast. In KRAS mutant colorectal cancer cell lines, genetic ablation of the human ortholog of IRE1, ERN1, does not affect growth but sensitizes to MEK inhibition. However, an ERN1 kinase inhibitor failed to show synergy with MEK inhibition, suggesting that a non-kinase function of ERN1 confers MEK inhibitor resistance. To investigate how ERN1 modulates MEK inhibitor responses, we performed genetic screens in ERN1 knockout KRAS mutant colon cancer cells to identify genes whose inactivation confers resistance to MEK inhibition. This genetic screen identified multiple negative regulators of JUN N-terminal kinase (JNK) /JUN signaling. Consistently, compounds targeting JNK/MAPK8 or TAK1/MAP3K7, which relay signals from ERN1 to JUN, display synergy with MEK inhibition. Conclusions We identify the ERN1-JNK-JUN pathway as a novel regulator of MEK inhibitor response in KRAS mutant colon cancer. The notion that multiple signaling pathways can activate JUN may explain why KRAS mutant tumor cells are traditionally seen as highly refractory to MEK inhibitor therapy. Our findings emphasize the need for the development of new therapeutics targeting JUN activating kinases, TAK1 and JNK, to sensitize KRAS mutant cancer cells to MEK inhibitors. Electronic supplementary material The online version of this article (10.1186/s13073-018-0600-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tonći Šuštić
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Sake van Wageningen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.,Department Genetics and Development, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, 10032, USA
| | - Evert Bosdriesz
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Robert J D Reid
- Department Genetics and Development, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, 10032, USA
| | - John Dittmar
- Department Genetics and Development, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, 10032, USA
| | - Cor Lieftink
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Roderick L Beijersbergen
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Lodewyk F A Wessels
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Rodney Rothstein
- Department Genetics and Development, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, 10032, USA.
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
| |
Collapse
|
27
|
Araya HF, Sepulveda H, Lizama CO, Vega OA, Jerez S, Briceño PF, Thaler R, Riester SM, Antonelli M, Salazar-Onfray F, Rodríguez JP, Moreno RD, Montecino M, Charbonneau M, Dubois CM, Stein GS, van Wijnen AJ, Galindo MA. Expression of the ectodomain-releasing protease ADAM17 is directly regulated by the osteosarcoma and bone-related transcription factor RUNX2. J Cell Biochem 2018; 119:8204-8219. [PMID: 29923217 DOI: 10.1002/jcb.26832] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/06/2018] [Indexed: 01/04/2023]
Abstract
Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.
Collapse
Affiliation(s)
- Héctor F Araya
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Hugo Sepulveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Carlos O Lizama
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar A Vega
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sofia Jerez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pedro F Briceño
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Marcelo Antonelli
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Rodríguez
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), University of Chile, Santiago, Chile
| | - Ricardo D Moreno
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Martine Charbonneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Claire M Dubois
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, The Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mario A Galindo
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
28
|
Liu X, Wang Y, Zhang L, Xu Z, Chu Q, Xu C, Sun Y, Gao Y. Combination of Runx2 and Cbfβ upregulates Amelotin gene expression in ameloblasts by directly interacting with cis‑enhancers during amelogenesis. Mol Med Rep 2018; 17:6068-6076. [PMID: 29436627 DOI: 10.3892/mmr.2018.8564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 01/05/2018] [Indexed: 11/05/2022] Open
Abstract
Amelotin (Amtn) is a recently identified enamel protein secreted by ameloblasts at late stage of enamel development. Runt‑related transcription factor 2 (Runx2) in combination with the coactivator core‑binding factor β (Cbfβ) regulates the early stages of tooth development. The aim of the present study was to investigate the role of Runx2 in the regulation of Amtn gene expression in ameloblasts. Immunohistochemistry was performed and the results revealed that Runx2 protein was predominantly expressed in the nuclei of ameloblasts during the transition stage and the maturation stage of enamel development, whereas Cbfβ was expressed in ameloblasts from the secretory stage to the maturation stage. Reverse transcription‑quantitative polymerase chain reaction results demonstrated that Runx2 knockdown decreased Amtn expression in ameloblast‑lineage cells and co‑expression of Runx2 and Cbfβ in ameloblast lineage cells induced an upregulation in Amtn gene expression. Two putative Runx2‑binding sites within the Amtn promoter were identified using bioinformatics analysis. Results of an electrophoretic mobility shift assay and chromatin immunoprecipitation indicated that Runx2/Cbfβ bound to specific DNA sequences. Site‑directed mutagenesis of the Runx2 binding sites within the Amtn promoter resulted in decreased basal promoter activity and did not affect the overexpressed Runx2/Cbfβ. The results of the present study suggest that Runx2 upregulates Amtn gene expression via binding directly to Runx2 sites within the Amtn promoter during amelogenesis.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Oral Biology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yumin Wang
- Department of Pediatric Dentistry, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Li Zhang
- Department of Pediatric Dentistry, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Zhenzhen Xu
- Department of Pediatric Dentistry, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Qing Chu
- Department of Pediatric Dentistry, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Chang Xu
- Department of Pediatric Dentistry, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Yan Sun
- Department of Oral Biology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Yuguang Gao
- Department of Oral Biology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
29
|
Komori T. Runx2, an inducer of osteoblast and chondrocyte differentiation. Histochem Cell Biol 2018; 149:313-323. [DOI: 10.1007/s00418-018-1640-6] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2018] [Indexed: 12/20/2022]
|
30
|
Abstract
Runx2 is the most upstream transcription factor essential for osteoblast differentiation. It regulates the expression of Sp7, the protein of which is a crucial transcription factor for osteoblast differentiation, as well as that of bone matrix genes including Spp1, Ibsp, and Bglap2. Runx2 is also required for chondrocyte maturation, and Runx3 has a redundant function with Runx2 in chondrocyte maturation. Runx2 regulates the expression of Col10a1, Spp1, Ibsp, and Mmp13 in chondrocytes. It also inhibits chondrocytes from acquiring the phenotypes of permanent cartilage chondrocytes. It regulates chondrocyte proliferation through the regulation of Ihh expression. Runx2 enhances osteoclastogenesis by regulating Rankl. Cbfb, which is a co-transcription factor for Runx family proteins, plays an important role in skeletal development by stabilizing Runx family proteins. In Cbfb isoforms, Cbfb1 is more potent than Cbfb2 in Runx2-dependent transcriptional regulation; however, the expression level of Cbfb2 is three-fold higher than that of Cbfb1, demonstrating the requirement of Cbfb2 in skeletal development. The expression of Runx2 in osteoblasts is regulated by a 343-bp enhancer located upstream of the P1 promoter. This enhancer is activated by an enhanceosome composed of Dlx5/6, Mef2, Tcf7, Ctnnb1, Sox5/6, Smad1, and Sp7. Thus, Runx2 is a multifunctional transcription factor that is essential for skeletal development, and Cbfb regulates skeletal development by modulating the stability and transcriptional activity of Runx family proteins.
Collapse
|
31
|
Jiang Q, Qin X, Kawane T, Komori H, Matsuo Y, Taniuchi I, Ito K, Izumi SI, Komori T. Cbfb2 Isoform Dominates More Potent Cbfb1 and Is Required for Skeletal Development. J Bone Miner Res 2016; 31:1391-404. [PMID: 26890219 DOI: 10.1002/jbmr.2814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 01/24/2016] [Accepted: 02/13/2016] [Indexed: 12/26/2022]
Abstract
Cbfb is a cotranscription factor that forms a heterodimer with Runx proteins Runx1, Runx2, and Runx3. It is required for fetal liver hematopoiesis and skeletal development. Cbfb has two functional isoforms, Cbfb1 and Cbfb2, which are formed by alternative splicing. To address the biological functions of these isoforms in skeletal development, we examined Cbfb1(-/-) and Cbfb2(-/-) mouse embryos. Intramembranous and endochondral ossification was retarded and chondrocyte and osteoblast differentiation was inhibited in Cbfb2(-/-) embryos but not in Cbfb1(-/-) embryos. Cbfb2 mRNA was upregulated in calvariae, limbs, livers, thymuses, and hearts of Cbfb1(-/-) embryos but Cbfb1 mRNA was not in those of Cbfb2(-/-) embryos, and the total amount of Cbfb1 and Cbfb2 mRNA in Cbfb1(-/-) embryos was similar to that in wild-type embryos but was severely reduced in Cbfb2(-/-) embryos. The absolute numbers of Cbfb2 mRNA in calvariae, limbs, livers, thymuses, and brains in wild-type embryos were about three times higher than those of Cbfb1 in the respective tissue. The levels of Runx proteins were reduced in calvariae, limbs, and primary osteoblasts from Cbfb2(-/-) embryos, but the reduction in Runx2 protein was very mild. Furthermore, the amounts of Runx proteins and Cbfb in Cbfb2(-/-) embryos differed similarly among skeletal tissues, livers, and thymuses, suggesting that Runx proteins and Cbfb are mutually required for their stability. Although Cbfb1(-/-) embryos developed normally, Cbfb1 induced chondrocyte and osteoblast differentiation and enhanced DNA binding of Runx2 more efficiently than Cbfb2. Our results indicate that modulations in the relative levels of the isoforms may adjust transcriptional activation by Runx2 to appropriate physiological levels. Cbfb2 was more abundant, but Cbfb1 was more potent for enhancing Runx2 activity. Although only Cbfb2 loss generated overt skeletal phenotypes, both may play major roles in skeletal development with functional redundancy. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Qing Jiang
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Xin Qin
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tetsuya Kawane
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hisato Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuki Matsuo
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shin-Ichi Izumi
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
32
|
Lim KE, Park NR, Che X, Han MS, Jeong JH, Kim SY, Park CY, Akiyama H, Kim JE, Ryoo HM, Stein JL, Lian JB, Stein GS, Choi JY. Core Binding Factor β of Osteoblasts Maintains Cortical Bone Mass via Stabilization of Runx2 in Mice. J Bone Miner Res 2016; 31:245. [PMID: 26804813 DOI: 10.1002/jbmr.2587] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
33
|
Deng P, Chen QM, Hong C, Wang CY. Histone methyltransferases and demethylases: regulators in balancing osteogenic and adipogenic differentiation of mesenchymal stem cells. Int J Oral Sci 2015; 7:197-204. [PMID: 26674421 PMCID: PMC5153596 DOI: 10.1038/ijos.2015.41] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2015] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are characterized by their self-renewing capacity and differentiation potential into multiple tissues. Thus, management of the differentiation capacities of MSCs is important for MSC-based regenerative medicine, such as craniofacial bone regeneration, and in new treatments for metabolic bone diseases, such as osteoporosis. In recent years, histone modification has been a growing topic in the field of MSC lineage specification, in which the Su(var)3–9, enhancer-of-zeste, trithorax (SET) domain-containing family and the Jumonji C (JmjC) domain-containing family represent the major histone lysine methyltransferases (KMTs) and histone lysine demethylases (KDMs), respectively. In this review, we summarize the current understanding of the epigenetic mechanisms by which SET domain-containing KMTs and JmjC domain-containing KDMs balance the osteogenic and adipogenic differentiation of MSCs.
Collapse
Affiliation(s)
- Peng Deng
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Qian-Ming Chen
- State Key Laboratory of Oral Disease, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Christine Hong
- Section of Orthodontics, School of Dentistry, University of California at Los Angeles, Los Angeles, USA
| | - Cun-Yu Wang
- Division of Oral Biology and Medicine, School of Dentistry, University of California at Los Angeles, Los Angeles, USA.,Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, USA
| |
Collapse
|
34
|
Fukuda T, Ochi H, Sunamura S, Haiden A, Bando W, Inose H, Okawa A, Asou Y, Takeda S. MicroRNA-145 regulates osteoblastic differentiation by targeting the transcription factor Cbfb. FEBS Lett 2015; 589:3302-8. [PMID: 26450370 DOI: 10.1016/j.febslet.2015.09.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 08/04/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023]
Abstract
Osteoblastic differentiation is regulated by various factors, including hormones and transcription factors. Runt-related transcription factor 2 (Runx2) is an essential player in osteoblastogenesis and transactivates its molecular target by creating a protein complex with its hetero-dimeric partner core binding factor beta (Cbfb). However, the molecular regulation of Cbfb expression remains unknown. Here, we identified miR-145 as a crucial regulator of Cbfb expression. The expression of miR-145 increased during osteoblastogenesis, indicating that miR-145 works as an inhibitor of osteoblastogenesis. Stable expression of miR-145 decreased endogenous Cbfb expression and inhibited osteoblastogenesis, in cooperation with miR-34c. Furthermore, miR-145 decreased bone regeneration in vivo. Our results indicate that miR-145 physiologically regulates osteoblast differentiation and bone formation via Cbfb expression by forming a regulatory microRNA network.
Collapse
Affiliation(s)
- Toru Fukuda
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hiroki Ochi
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Satoko Sunamura
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Akina Haiden
- Department of Emergency & Critical Care Medicine, School of Medicine, Keio University, Shinanomachi 35, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Waka Bando
- Section of Nephrology, Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, Keio University, Shinanomachi 35, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Hiroyuki Inose
- Department of Orthopedic Surgery, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Atsushi Okawa
- Department of Orthopedic Surgery and Global Center of Excellence (GCOE) Program, International Research Center for Molecular Science in Tooth and Bone Diseases, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Yoshinori Asou
- Department of Orthopedic Surgery and Global Center of Excellence (GCOE) Program, International Research Center for Molecular Science in Tooth and Bone Diseases, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Shu Takeda
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
35
|
Gou Y, Zhang T, Xu J. Transcription Factors in Craniofacial Development: From Receptor Signaling to Transcriptional and Epigenetic Regulation. Curr Top Dev Biol 2015; 115:377-410. [PMID: 26589933 DOI: 10.1016/bs.ctdb.2015.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Craniofacial morphogenesis is driven by spatial-temporal terrains of gene expression, which give rise to stereotypical pattern formation. Transcription factors are key cellular components that control these gene expressions. They are information hubs that integrate inputs from extracellular factors and environmental cues, direct epigenetic modifications, and define transcriptional status. These activities allow transcription factors to confer specificity and potency to transcription regulation during development.
Collapse
Affiliation(s)
- Yongchao Gou
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA
| | - Tingwei Zhang
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA; State Key Laboratory of Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Xu
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA.
| |
Collapse
|
36
|
Park NR, Lim KE, Han MS, Che X, Park CY, Kim JE, Taniuchi I, Bae SC, Choi JY. Core Binding Factor β Plays a Critical Role During Chondrocyte Differentiation. J Cell Physiol 2015; 231:162-71. [DOI: 10.1002/jcp.25068] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/05/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Na-Rae Park
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Kyung-Eun Lim
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Min-Su Han
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Xiangguo Che
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Clara Yongjoo Park
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine; Kyungpook National University School of Medicine; Daegu Republic of Korea
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation; RIKEN Research Center for Allergy and Immunology; Kanagawa Japan
| | - Suk-Chul Bae
- Department of Biochemistry; Institute for Tumor Research; Chungbuk National University; College of Medicine; Cheongju Republic of Korea
| | - Je-Yong Choi
- Department of Biochemistry and Cell Biology; Skeletal Diseases Genome Research Center; Cell and Matrix Research Institute; BK21 Plus KNU Biomedical Convergence Program; Kyungpook National University School of Medicine; Daegu Republic of Korea
| |
Collapse
|
37
|
Bauer O, Sharir A, Kimura A, Hantisteanu S, Takeda S, Groner Y. Loss of osteoblast Runx3 produces severe congenital osteopenia. Mol Cell Biol 2015; 35:1097-109. [PMID: 25605327 PMCID: PMC4355527 DOI: 10.1128/mcb.01106-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/07/2014] [Accepted: 12/30/2014] [Indexed: 11/20/2022] Open
Abstract
Congenital osteopenia is a bone demineralization condition that is associated with elevated fracture risk in human infants. Here we show that Runx3, like Runx2, is expressed in precommitted embryonic osteoblasts and that Runx3-deficient mice develop severe congenital osteopenia. Runx3-deficient osteoblast-specific (Runx3(fl/fl)/Col1α1-cre), but not chondrocyte-specific (Runx3(fl/fl)/Col1α2-cre), mice are osteopenic. This demonstrates that an osteoblastic cell-autonomous function of Runx3 is required for proper osteogenesis. Bone histomorphometry revealed that decreased osteoblast numbers and reduced mineral deposition capacity in Runx3-deficient mice cause this bone formation deficiency. Neonatal bone and cultured primary osteoblast analyses revealed a Runx3-deficiency-associated decrease in the number of active osteoblasts resulting from diminished proliferation and not from enhanced osteoblast apoptosis. These findings are supported by Runx3-null culture transcriptome analyses showing significant decreases in the levels of osteoblastic markers and increases in the levels of Notch signaling components. Thus, while Runx2 is mandatory for the osteoblastic lineage commitment, Runx3 is nonredundantly required for the proliferation of these precommitted cells, to generate adequate numbers of active osteoblasts. Human RUNX3 resides on chromosome 1p36, a region that is associated with osteoporosis. Therefore, RUNX3 might also be involved in human bone mineralization.
Collapse
Affiliation(s)
- Omri Bauer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Amnon Sharir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ayako Kimura
- Department of Orthopedics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shay Hantisteanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shu Takeda
- Department of Orthopedics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|