Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Mol Cancer Ther ; 21(2): 347-358, 2022 02.
Article in English | MEDLINE | ID: mdl-34907087

ABSTRACT

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM). The proteasome inhibitor bortezomib is one of the most effective first-line chemotherapeutic drugs for multiple myeloma; however, 15% to 20% of high-risk patients do not respond to or become resistant to this drug and the mechanisms of chemoresistance remain unclear. We previously demonstrated that multiple myeloma cells inhibit Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB), and that this OB-Runx2 deficiency induces a cytokine-rich and immunosuppressive microenvironment in the BM. In the current study, we assessed the impact of OB-Runx2 deficiency on the outcome of bortezomib treatment using OB-Runx2+/+ and OB-Runx2-/- mouse models of multiple myeloma. In vitro and in vivo experiments revealed that OB-Runx2 deficiency induces multiple myeloma cell resistance to bortezomib via the upregulation of immunosuppressive myeloid-derived suppressor cells (MDSCs), downregulation of cytotoxic T cells, and activation of TGFß1 in the BM. In multiple myeloma tumor-bearing OB-Runx2-/- mice, treatment with SRI31277, an antagonist of thrombospondin-1 (TSP-1)-mediated TGFß1 activation, reversed the BM immunosuppression and significantly reduced tumor burden. Furthermore, treatment with SRI31277 combined with bortezomib alleviated multiple myeloma cell resistance to bortezomib-induced apoptosis caused by OB-Runx2 deficiency in cocultured cells and produced a synergistic effect on tumor burden in OB-Runx2-/- mice. Depletion of MDSCs by 5-fluorouracil or gemcitabine similarly reversed the immunosuppressive effects and bortezomib resistance induced by OB-Runx2 deficiency in tumor-bearing mice, indicating the importance of the immune environment for drug resistance and suggesting new strategies to overcome bortezomib resistance in the treatment of multiple myeloma.


Subject(s)
Bone Marrow/metabolism , Bortezomib/therapeutic use , Core Binding Factor Alpha 1 Subunit/deficiency , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Osteoblasts/metabolism , Thrombospondin 1/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Bortezomib/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Multiple Myeloma/pathology
2.
J Mol Histol ; 52(3): 545-553, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33763807

ABSTRACT

Junctional epithelium (JE) attaching to the enamel surface seals gaps around the teeth, functioning as the first line of gingival defense. Runt-related transcription factor 2 (Runx2) plays a role in epithelial cell fate, and the deficiency of Runx2 in JE causes periodontal destruction, while its effect on the barrier function of JE remains largely unexplored. In the present study, hematoxylin-eosin (H&E) staining revealed the morphological differences of JE between wild-type (WT) and Runx2 conditional knockout (cKO) mice. We speculated that these changes were related to the down-regulation of E-cadherin (E-cad), junctional adhesion molecule 1 (JAM1), and integrin ß6 (ITGB6) in JE. Moreover, immunohistochemistry (IHC) was conducted to assess the expressions of these proteins. To verify the relationship between Runx2 and the three above-mentioned proteins, human gingival epithelial cells (HGEs) were cultured for in vitro experiment. The expression of Runx2 in HEGs was depleted by lentivirus. Quantitative real-time PCR (qRT-PCR) and Western blotting analysis were adopted to analyze the differences in mRNA and protein expressions. Taken together, Runx2 played a crucial role in maintaining the structure and function integrality of JE via regulating the expressions of E-cad and JAM1.


Subject(s)
Cadherins/metabolism , Core Binding Factor Alpha 1 Subunit/deficiency , Epithelium/metabolism , Junctional Adhesion Molecules/metabolism , Molar/metabolism , Animals , Core Binding Factor Alpha 1 Subunit/metabolism , Down-Regulation , Epithelial Cells/metabolism , Gingiva/cytology , Humans , Integrin beta Chains/metabolism , Mandible/metabolism , Mice, Knockout , Periodontium/metabolism
3.
Cancer Genomics Proteomics ; 17(2): 161-168, 2020.
Article in English | MEDLINE | ID: mdl-32108038

ABSTRACT

BACKGROUND/AIM: Osteoblastoma is a rare benign tumor of the bones in which recurrent rearrangements of FOS have been found. Our aim was to investigate two osteoblastomas for possible genetic aberrations. MATERIALS AND METHODS: Cytogenetic, RNA sequencing, and molecular analyses were performed. RESULTS: A FOS-ANKH transcript was found in the first tumor, whereas a FOS-RUNX2 was detected in the second. Exon 4 of FOS fused with sequences either from intron 1 of ANKH or intron 5 of RUNX2. The fusion events introduced a stop codon and removed sequences involved in the regulation of FOS. CONCLUSION: Rearrangements and fusions of FOS show similarities with those of HMGA2 (a feature of leiomyomas and lipomas) and CSF1 (tenosynovial giant cell tumors). The replacement of a 3'-untranslated region, controlling the gene's expression, by a new sequence is thus a common pathogenetic theme shared by FOS, HMGA2, and CSF1 in many benign connective tissue tumors.


Subject(s)
Bone Neoplasms/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Osteoblastoma/genetics , Phosphate Transport Proteins/genetics , Base Sequence , Bone Neoplasms/metabolism , Child , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Gene Expression , Humans , Karyotype , Male , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Osteoblastoma/metabolism , Phosphate Transport Proteins/metabolism
4.
Cancer Res ; 80(5): 1036-1048, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31911552

ABSTRACT

Multiple myeloma is a plasma cell malignancy that thrives in the bone marrow (BM), with frequent progression to new local and distant bone sites. Our previous studies demonstrated that multiple myeloma cells at primary sites secrete soluble factors and suppress osteoblastogenesis via the inhibition of Runt-related transcription factor 2 (Runx2) in pre- and immature osteoblasts (OB) in new bone sites, prior to the arrival of metastatic tumor cells. However, it is unknown whether OB-Runx2 suppression in new bone sites feeds back to promote multiple myeloma dissemination to and progression in these areas. Hence, we developed a syngeneic mouse model of multiple myeloma in which Runx2 is specifically deleted in the immature OBs of C57BL6/KaLwRij mice (OB-Runx2-/- mice) to study the effect of OB-Runx2 deficiency on multiple myeloma progression in new bone sites. In vivo studies with this model demonstrated that OB-Runx2 deficiency attracts multiple myeloma cells and promotes multiple myeloma tumor growth in bone. Mechanistic studies further revealed that OB-Runx2 deficiency induces an immunosuppressive microenvironment in BM that is marked by an increase in the concentration and activation of myeloid-derived suppressor cells (MDSC) and the suppression and exhaustion of cytotoxic CD8+ T cells. In contrast, MDSC depletion by either gemcitabine or 5-fluorouracil treatment in OB-Runx2-/- mice prevented these effects and inhibited multiple myeloma tumor growth in BM. These novel discoveries demonstrate that OB-Runx2 deficiency in new bone sites promotes multiple myeloma dissemination and progression by increasing metastatic cytokines and MDSCs in BM and inhibiting BM immunity. Importantly, MDSC depletion can block multiple myeloma progression promoted by OB-Runx2 deficiency.Significance: This study demonstrates that Runx2 deficiency in immature osteoblasts at distant bone sites attracts myeloma cells and allows myeloma progression in new bone sites via OB-secreted metastatic cytokines and MDSC-mediated suppression of bone marrow immunity.


Subject(s)
Bone Neoplasms/secondary , Core Binding Factor Alpha 1 Subunit/deficiency , Multiple Myeloma/pathology , Osteoblasts/pathology , Tumor Microenvironment/immunology , Animals , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow/pathology , Bone Neoplasms/immunology , Bone and Bones/cytology , Bone and Bones/drug effects , Bone and Bones/immunology , Bone and Bones/pathology , Cell Line, Tumor/transplantation , Core Binding Factor Alpha 1 Subunit/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Disease Models, Animal , Female , Fluorouracil , Humans , Male , Mice , Mice, Knockout , Multiple Myeloma/immunology , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/pathology , Osteoblasts/immunology , Gemcitabine
5.
Sci Rep ; 9(1): 15596, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31666602

ABSTRACT

Notch signaling is involved in the early onset of osteoarthritis. The aim of this study was to investigate the role of Notch signaling changes during proliferation and differentiation of chondrocyte, and to testify the mechanism of MMP-13 regulation by Notch and Runx2 expression changes during osteoarthritis. In this study, Chondrocytes were isolated from rat knee cartilages. Notch signaling was activated/inhibited by Jagged-1/DAPT. Proliferative capacity of Chondrocytes was analyzed by CCK-8 staining and EdU labeling. ColX, Runx2 and MMP-13 expressions were analyzed as cell differentiation makers. Then, Runx2 gene expression was interfered using lentivirus transfection (RNAi) and was over-expressed by plasmids transfected siRNA in chondrocytes, and MMP-13 expression was analyzed after Jagged-1/DAPT treatment. In vivo, an intra-articular injection of shRunx2 lentivirus followed with Jagged1/DAPT treatments was performed in rats. MMP-13 expression in articular cartilage was detected by immunohistochemistry. Finally, MMP-13 expression changes were analyzed in chondrocytes under IL-1ß stimulation. Our findings showed that, CCK-8 staining and EdU labeling revealed suppression of cell proliferation by Notch signaling activation after Jagged-1 treatment in chondrocytes. Promoted differentiation was also observed, characterized by increased expressions of Col X, MMP-13 and Runx2. Meanwhile, Sox9, aggrecan and Col II expressions were down-regulated. The opposite results were observed in Notch signaling inhibited cells by DAPT treatment. In addition, Runx2 RNAi significantly attenuated the 'regulatory sensitivity' of Notch signaling on MMP-13 expression both in vitro and in vivo. However, we found there wasn't significant changes of this 'regulatory sensitivity' of Notch signaling after Runx2 over-expression. Under IL-1ß circumstance, MMP-13 expression could be reduced by both DAPT treatment and Runx2 RNAi, while Runx2 interference also attenuated the 'regulatory sensitivity' of Notch in MMP-13 under IL-1ß stimulation. In conclusion, Notch signaling is an important regulator on rat chondrocyte proliferation and differentiation, and this regulatory effect was partially mediated by proper Runx2 expression under both normal and IL-1ß circumstances. In the meanwhile, DAPT treatment could effectively suppress expression of MMP-13 stimulated by IL-1 ß.


Subject(s)
Chondrocytes/cytology , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Developmental , Matrix Metalloproteinase 13/genetics , Receptors, Notch/metabolism , Signal Transduction , Animals , Cartilage, Articular/cytology , Cell Differentiation , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/genetics , Gene Knockdown Techniques , Male , Rats , Rats, Sprague-Dawley
6.
J Cell Physiol ; 234(4): 3436-3444, 2019 04.
Article in English | MEDLINE | ID: mdl-30387127

ABSTRACT

Runt-related transcription factor-2 (Runx2) is essential for chondrocyte maturation during cartilage development and embryonic mandibular condylar development. The process that chondrocytes, especially a subgroup of hypertrophic chondrocytes (HC), could transform into bone cells in mandibular condyle growth makes chondrocytes crucially important for normal endochondral bone formation. To determine whether Runx2 regulates postnatal condylar cartilage growth and tissue homeostasis, we deleted Runx2 in chondrocytes in postnatal mice and assessed the consequences on temporomandibular joint (TMJ) cartilage growth and remodeling. The cell lineage tracing data provide information demonstrating the role of chondrocytes in subchondral bone remodeling. The histologic and immunohistochemical data showed that Runx2 deficiency caused condylar tissue disorganization, including loss of HC and reduced hypertrophic zone, reduced proliferative chondrocytes, and decreased cartilage matrix production. Expression of Col10a1, Mmp13, Col2a1, Aggrecan, and Ihh was significantly reduced in Runx2 knockout mice. The findings of this study demonstrate that Runx2 is required for chondrocyte proliferation and hypertrophy in TMJ cartilage and postnatal TMJ cartilage growth and homeostasis, and that Runx2 may play an important role in regulation of chondrocyte-derived subchondral bone remodeling.


Subject(s)
Cell Proliferation , Chondrocytes/metabolism , Chondrogenesis , Core Binding Factor Alpha 1 Subunit/deficiency , Mandibular Condyle/metabolism , Temporomandibular Joint/metabolism , Animals , Bone Remodeling , Cell Lineage , Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Genotype , Homeostasis , Hypertrophy , Mandibular Condyle/pathology , Mice, Knockout , Phenotype , Temporomandibular Joint/pathology
7.
Sci Rep ; 8(1): 9594, 2018 06 25.
Article in English | MEDLINE | ID: mdl-29941908

ABSTRACT

Runt-related transcription factor 2 (Runx2) is involved in the early stage of tooth development. However, only few studies have reported the role of Runx2 in enamel development, which may be attributed to that Runx2 full knockout mice cannot survive after birth. In the present study, we successfully established a Runx2-deficient mouse model using a conditional knockout (cKO) method. We observed a significant reduction in the degree of mineralization and the decreased size of enamel rods in cKO mice. Histological analysis showed the retained enamel proteins in enamel layer at maturation stage in cKO molars. Further analysis by qRT-PCR revealed that the expressions of genes encoding enamel structure proteins, such as amelogenin (AMELX), ameloblastin (AMBN) and enamelin (ENAM), were increased in cKO enamel organs. On the other hand, the expression of kallikrein-related peptidase-4 (KLK4) at the mRNA and protein levels was dramatically decreased from late secretory stage to maturation stage in cKO enamel organs, while the expression of matrix metalloproteinase-20 (MMP-20) was not significantly altered. Finally, immunohistochemistry indicated that the uptake of amelogenins by ameloblasts was significantly decreased in cKO mice. Taken together, Runx2 played critical roles in controlling enamel maturation by increasing synthesis of KLK4 and decreasing synthesis of AMELX, AMBN and ENAM.


Subject(s)
Ameloblasts/metabolism , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/genetics , Dental Enamel/cytology , Dental Enamel/growth & development , Gene Knockout Techniques , Amelogenin/metabolism , Animals , Dental Enamel/metabolism , Dental Enamel Proteins/metabolism , Gene Expression Regulation , Kallikreins/metabolism , Mice , Minerals/metabolism
8.
Int J Mol Sci ; 19(1)2018 Jan 06.
Article in English | MEDLINE | ID: mdl-29316655

ABSTRACT

Neural EGFL like 1 (Nell-1) is essential for chondrogenic differentiation, maturation, and regeneration. Our previous studies have demonstrated that Nell-1's pro-chondrogenic activities are predominantly reliant upon runt-related transcription factor 3 (Runx3)-mediated Indian hedgehog (Ihh) signaling. Here, we identify the nuclear factor of activated T-cells 1 (Nfatc1) as the key transcriptional factor mediating the Nell-1 → Runx3 signal transduction in chondrocytes. Using chromatin immunoprecipitation assay, we were able to determine that Nfatc1 binds to the -833--810 region of the Runx3-promoter in response to Nell-1 treatment. By revealing the Nell-1 → Nfatc1 → Runx3 → Ihh cascade, we demonstrate the involvement of Nfatc1, a nuclear factor of activated T-cells, in chondrogenesis, while providing innovative insights into developing a novel therapeutic strategy for cartilage regeneration and other chondrogenesis-related conditions.


Subject(s)
Calcium-Binding Proteins/pharmacology , Core Binding Factor Alpha 3 Subunit/metabolism , Glycoproteins/pharmacology , NFATC Transcription Factors/metabolism , Up-Regulation/drug effects , Adipose Tissue/cytology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/genetics , Glycoproteins/genetics , Glycoproteins/metabolism , Mice , Mice, Knockout , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
9.
Clin Cancer Res ; 24(4): 834-846, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29167276

ABSTRACT

Purpose: Intratumoral androgen synthesis (IAS) is a key mechanism promoting androgen receptor (AR) reactivation and antiandrogen resistance in castration-resistant prostate cancer (CRPC). However, signaling pathways driving aberrant IAS remain poorly understood.Experimental Design: The effect of components of the AKT-RUNX2-osteocalcin (OCN)-GPRC6A-CREB signaling axis on expression of steroidogenesis genes CYP11A1 and CYP17A1 and testosterone level were examined in PTEN-null human prostate cancer cell lines. Pten knockout mice were used to examine the effect of Runx2 heterozygous deletion or abiraterone acetate (ABA), a prodrug of the CYP17A1 inhibitor abiraterone on Cyp11a1 and Cyp17a1 expression, testosterone level and tumor microenvironment (TME) remodeling in vivoResults: We uncovered that activation of the AKT-RUNX2-OCN-GPRC6A-CREB signaling axis induced expression of CYP11A1 and CYP17A1 and testosterone production in PTEN-null prostate cancer cell lines in culture. Deletion of Runx2 in Pten homozygous knockout prostate tumors decreased Cyp11a1 and Cyp17a1 expression, testosterone level, and tumor growth in castrated mice. ABA treatment also inhibited testosterone synthesis and alleviated Pten loss-induced tumorigenesis in vivoPten deletion induced TME remodeling, but Runx2 heterozygous deletion or ABA treatment reversed the effect of Pten loss by decreasing expression of the collagenase Mmp9.Conclusions: Abnormal RUNX2 activation plays a pivotal role in PTEN loss-induced IAS and TME remodeling, suggesting that the identified signaling cascade represents a viable target for effective treatment of PTEN-null prostate cancer, including CRPC. Clin Cancer Res; 24(4); 834-46. ©2017 AACR.


Subject(s)
Abiraterone Acetate/pharmacology , Androgens/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Tumor Microenvironment/drug effects , Animals , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/deficiency , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Knockout , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Steroid 17-alpha-Hydroxylase/genetics , Steroid 17-alpha-Hydroxylase/metabolism , Testosterone/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
10.
Am J Pathol ; 188(2): 392-403, 2018 02.
Article in English | MEDLINE | ID: mdl-29137952

ABSTRACT

The pro-chondrogenic function of runt-related transcription factor 2 (Runx2) was previously considered to be dependent on direct binding with the promoter of Indian hedgehog (Ihh)-the major regulator of chondrocyte differentiation, proliferation, and maturation. The authors' previous studies identified neural EGFL like 1 (Nell-1) as a Runx2-responsive growth factor for chondrogenic differentiation and maturation. In this study, it was further revealed that the pro-chondrogenic activities of Nell-1 also rely on Ihh signaling, by showing: i) Nell-1 significantly elevated Ihh signal transduction; ii) Nell-1 deficiency markedly reduced Ihh activation in chondrocytes; and iii) Nell-1-stimulated chondrogenesis was significantly reduced by the specific hedgehog inhibitor cyclopamine. Importantly, the authors demonstrated that Nell-1-responsive Ihh signaling and chondrogenic differentiation extended to Runx2-/- models in vitro and in vivo. In Runx2-/- chondrocytes, Nell-1 stimulated the expression and signal transduction of Runx3, another transcription factor required for complete chondrogenic differentiation and maturation. Furthermore, knocking down Runx3 in Runx2-/- chondrocytes abolished Nell-1's stimulation of Ihh-associated molecule expression, which validates Runx3 as a major mediator of Nell-1-stimulated Ihh activation. For the first time, the Runx2→Nell-1→Runx3→Ihh signaling cascade during chondrogenic differentiation and maturation has been identified as an alternative, but critical, pathway for Runx2 to function as a pro-chondrogenic molecule via Nell-1.


Subject(s)
Calcium-Binding Proteins/physiology , Chondrocytes/physiology , Core Binding Factor Alpha 1 Subunit/physiology , Glycoproteins/physiology , Hedgehog Proteins/physiology , Animals , Cartilage/cytology , Cartilage/physiology , Cell Differentiation/physiology , Cells, Cultured , Chondrocytes/cytology , Chondrogenesis/physiology , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 3 Subunit/physiology , Mice, Knockout , Signal Transduction/physiology
11.
Biomater Sci ; 5(9): 1910-1921, 2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28722044

ABSTRACT

In the context of regenerative medicine, the use of RNA interference mechanisms has already proven its efficiency in targeting specific gene expression with the aim of enhancing, accelerating or, more generally, directing stem cell differentiation. However, achievement of good transfection levels requires the use of a gene vector. For in vivo applications, synthetic vectors are an interesting option to avoid possible issues associated with viral vectors (safety, production costs, etc.). Herein, we report on the design of tripartite polyionic complex micelles as original non-viral polymeric vectors suited for mesenchymal stem cell transfection with siRNA. Three micelle formulations were designed to exhibit pH-triggered disassembly in an acidic pH range comparable to that of endosomes. One formulation was selected as the most promising with the highest siRNA loading capacity while clearly maintaining pH-triggered disassembly properties. A thorough investigation of the internalization pathway of micelles into cells with tagged siRNA was made before showing an efficient inhibition of Runx2 expression in primary bone marrow-derived stem cells. This work evidenced PIC micelles as promising synthetic vectors that allow efficient MSC transfection and control over their behavior, from the perspective of their clinical use.


Subject(s)
Drug Carriers/chemistry , Mesenchymal Stem Cells/metabolism , Micelles , RNA, Small Interfering/genetics , Transfection/methods , Animals , Base Sequence , Cell Survival/drug effects , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/genetics , Drug Carriers/metabolism , Drug Carriers/toxicity , Endocytosis , Mice
12.
J Cell Physiol ; 232(1): 182-91, 2017 01.
Article in English | MEDLINE | ID: mdl-27064596

ABSTRACT

DICER is the central enzyme that cleaves precursor microRNAs (miRNAs) into 21-25 nucleotide duplex in cell lineage differentiation, identity, and survival. In the current study, we characterized the specific bone metabolism genes and corresponding miRNAs and found that DICER and Runt-related transcription factor 2 (Runx2) expressions increased simultaneously during osteogenic differentiation. Luciferase assay showed that Runx2 significantly increased the expression levels of DICER luciferase promoter reporter. Our analysis also revealed weaker DICER expression in embryos of Runx2 knock out mice (Runx2 -/-) compared with that of Runx2 +/- and Runx2 +/+ mice. We further established the calvarial bone critical-size defect (CSD) mouse model. The bone marrow stromal cells (BMSCs) transfected with siRNA targeting DICER were combined with silk scaffolds and transplanted into calvarial bone CSDs. Five weeks post-surgery, micro-CT analysis revealed impaired bone formation, and repairing in calvarial defects with the siRNA targeting DICER group. In conclusion, our results suggest that DICER is specifically regulated by osteogenic master gene Runx2 that binds to the DICER promoter. Consequently, DICER cleaves precursors of miR-335-5p and miR-17-92 cluster to form mature miRNAs, which target and decrease the Dickkopf-related protein 1 (DKK1), and proapoptotic factor BIM levels, respectively, leading to an enhanced Wnt/ß-catenin signaling pathway. These intriguing results reveal a central mechanism underlying lineage-specific regulation by a Runx2/DICER/miRNAs cascade during osteogenic differentiation and bone development. Our study, also suggests a potential application of modulating DICER expression for bone tissue repair and regeneration. J. Cell. Physiol. 232: 182-191, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , DEAD-box RNA Helicases/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Ribonuclease III/metabolism , Animals , Cell Line , Core Binding Factor Alpha 1 Subunit/deficiency , DEAD-box RNA Helicases/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Osteoblasts/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Ribonuclease III/genetics
13.
J Dent Res ; 94(8): 1113-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25916343

ABSTRACT

Tooth organogenesis depends on genetically programmed sequential and reciprocal inductive interactions between the dental epithelium and neural crest-derived mesenchyme. Previous studies showed that the Msx1 and Runx2 transcription factors are required for activation of odontogenic signals, including Bmp4 and Fgf3, in the early tooth mesenchyme to drive tooth morphogenesis through the bud-to-cap transition and that Runx2 acts downstream of Msx1 to activate Fgf3 expression. Recent studies identified Osr2 as a repressor of tooth development and showed that inactivation of Osr2 rescued molar tooth morphogenesis in the Msx1(-/-) mutant mice as well as in mice with neural crest-specific inactivation of Bmp4. Here we show that Runx2 expression is expanded in the tooth bud mesenchyme in Osr2(-/-) mutant mouse embryos and is partially restored in the tooth mesenchyme in Msx1(-/-)Osr2(-/-) mutants in comparison with Msx1(-/-) and wild-type embryos. Whereas mandibular molar development arrested at the bud stage and maxillary molar development arrested at the bud-to-cap transition in Runx2(-/-) mutant mice, both mandibular and maxillary molar tooth germs progressed to the early bell stage, with rescued expression of Msx1 and Bmp4 in the dental papilla as well as expression of Bmp4, p21, and Shh in the primary enamel knot in the Osr2(-/-)Runx2(-/-) compound mutants. In contrast to the Msx1(-/-)Osr2(-/-) compound mutants, which exhibit nearly normal first molar morphogenesis, the Osr2(-/-)Runx2(-/-) compound mutant embryos failed to activate the expression of Fgf3 and Fgf10 in the dental papilla and exhibited significant deficit in cell proliferation in both the dental epithelium and mesenchyme in comparison with the control embryos. These data indicate that Runx2 synergizes with Msx1 to drive tooth morphogenesis through the bud-to-cap transition and that Runx2 controls continued tooth growth and morphogenesis beyond the cap stage through activation of Fgf3 and Fgf10 expression in the dental papilla.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Odontogenesis/physiology , Tooth/embryology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Cell Death , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/deficiency , Female , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factor 3/metabolism , In Situ Nick-End Labeling , MSX1 Transcription Factor/metabolism , Mice , Pregnancy , Tooth/metabolism
14.
J Bone Miner Res ; 30(1): 71-82, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25079226

ABSTRACT

The Runx2 transcription factor is critical for commitment to the osteoblast lineage. However, its role in committed osteoblasts and its functions during postnatal skeletogenesis remain unclear. We established a Runx2-floxed line with insertion of loxP sites around exon 8 of the Runx2 gene. The Runx2 protein lacking the region encoded by exon 8 is imported into the nucleus and binds target DNA but exhibits diminished transcriptional activity. We specifically deleted the Runx2 gene in committed osteoblasts using 2.3-kb col1a-Cre transgenic mice. Surprisingly, the homozygous Runx2 mutant mice were born alive. The Runx2 heterozygous and homozygous null were grossly indistinguishable from wild-type littermates at birth. Runx2 deficiency did not alter proliferative capacity of osteoblasts during embryonic development (E18). Chondrocyte differentiation and cartilage growth in mutants was similar to wild-type mice from birth to 3 months of age. Analysis of the embryonic skeleton revealed poor calcification in homozygous mutants, which was more evident in bones formed by intramembranous ossification. Runx2 mutants showed progressive retardation in postnatal growth and exhibited significantly low bone mass by 1 month of age. Decreased bone formation was associated with decreased gene expression of osteoblast markers and impaired collagen assembly in the extracellular matrix. Consequently, Runx2 mutant bones exhibited decreased stiffness and structural integrity. By 3 months of age, bone acquisition in mutant mice was roughly half that of wild-type littermates. In addition to impaired osteoblast function, mutant mice showed markedly decreased osteoclast number and postnatal bone resorption. Taken together, functional deficiency of Runx2 in osteoblasts does not result in failed embryonic skeletogenesis but disrupts postnatal bone formation.


Subject(s)
Bone Development , Bone Resorption/metabolism , Cell Differentiation , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/deficiency , Osteoblasts/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Gene Expression Regulation/genetics , Homozygote , Mice , Mice, Knockout , Osteoblasts/pathology
17.
Int J Mol Sci ; 14(7): 14321-32, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23846726

ABSTRACT

Thrombospondin-1 (TSP-1), a matricellular protein widely acclaimed to be involved in the inhibition of angiogenesis and tumorigenesis, is synthesized and secreted by many cell types, including osteoblast and cancer cells. TSP-1 is highly upregulated during early stage of osteogenesis, whereas it inhibits terminal osteoblast differentiation. Expression of TSP-1 is downregulated in cancer cells, and its ectopic expression has been shown to restrain tumor growth. Transcriptional regulation of TSP-1 in osteogenesis and cancer is poorly understood; this prompted us to study its regulation by the two key regulators of the aforementioned processes: Runx2 and Runx3. Through a PCR-based cDNA subtraction technique, we identified and cloned a cDNA fragment for mouse TSP-1, whose expression was dramatically upregulated in response to Runx2 expression in mesenchymal stem cells. Moreover, TSP-1 expression was considerably reduced in the lung of Runx2 knockout mouse. On the other hand, TSP-1 gene expression drastically increased at both the transcriptional and translational levels in response to Runx3 expression in B16-F10 melanoma cells. In line with this, Runx2 and Runx3 bound to the TSP-1 promoter and stimulated its activity. Hence, these results provide first line of evidence that TSP-1 is a transcriptional target gene of Runx2 and Runx3.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 3 Subunit/metabolism , Thrombospondin 1/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Lung/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/metabolism , Transcription, Genetic
18.
J Bone Miner Res ; 28(10): 2064-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23553905

ABSTRACT

Global gene deletion studies in mice and humans have established the pivotal role of runt related transcription factor-2 (Runx2) in both intramembranous and endochondral ossification processes during skeletogenesis. In this study, we for the first time generated mice carrying a conditional Runx2 allele with exon 4, which encodes the Runt domain, flanked by loxP sites. These mice were crossed with α1(I)-collagen-Cre or α1(II)-collagen-Cre transgenic mice to obtain osteoblast-specific or chondrocyte-specific Runx2 deficient mice, respectively. As seen in Runx2(-/-) mice, perinatal lethality was observed in α1(II)-Cre;Runx2(flox/flox) mice, but this was not the case in animals in which α1(I)-collagen-Cre was used to delete Runx2. When using double-staining with Alizarin red for mineralized matrix and Alcian blue for cartilaginous matrix, we observed previously that mineralization was totally absent at embryonic day 15.5 (E15.5) throughout the body in Runx2(-/-) mice, but was found in areas undergoing intramembranous ossification such as skull and clavicles in α1(II)-Cre;Runx2(flox/flox) mice. In newborn α1(II)-Cre;Runx2(flox/flox) mice, mineralization impairment was restricted to skeletal areas undergoing endochondral ossification including long bones and vertebrae. In contrast, no apparent skeletal abnormalities were seen in mutant embryo, newborn, and 3-week-old to 6-week old-mice in which Runx2 had been deleted with the α1(I)-collagen-Cre driver. These results suggest that Runx2 is absolutely required for endochondral ossification during embryonic and postnatal skeletogenesis, but that disrupting its expression in already committed osteoblasts as achieved here with the α1(I)-collagen-Cre driver does not affect overtly intramembranous and endochondral ossification. The Runx2 floxed allele established here is undoubtedly useful for investigating the role of Runx2 in particular cells.


Subject(s)
Bone Development , Chondrocytes/metabolism , Core Binding Factor Alpha 1 Subunit/deficiency , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Animals , Female , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
19.
PLoS One ; 8(1): e54317, 2013.
Article in English | MEDLINE | ID: mdl-23372705

ABSTRACT

Runx2, a member of the family of runt-related transcription factors, is rhythmically expressed in bone and may be involved in circadian rhythms in bone homeostasis and osteogenesis. Runx2 is also expressed in the brain, but its function is unknown. We tested the hypothesis that in the brain, Runx2 may interact with clock-controlled genes to regulate circadian rhythms in behavior. First, we demonstrated diurnal and circadian rhythms in the expression of Runx2 in the mouse brain. Expression of Runx2 mRNA and protein mirrored that of the core clock genes, Period1 and Period2, in the suprachiasmatic nucleus (SCN), the paraventricular nucleus and the olfactory bulb. The rhythm of Runx2 expression was eliminated in the SCN of Bmal1(-/-) mice. Moreover, by crossbreeding mPer2(Luc) mice with Runx2(+/-) mice and recording bioluminescence rhythms, a significant lengthening of the period of rhythms was detected in cultured SCN of Runx2(-/-) animals compared to either Runx2(+/-) or Runx2(+/+) mice. Behavioral analyses of Runx2 mutant mice revealed that Runx2(+/-) animals displayed a significantly lengthened free-running period of running wheel activity compared to Runx2(+/+) littermates. Taken together, these findings provide evidence for clock gene-mediated rhythmic expression of Runx2, and its functional role in regulating circadian period at the level of the SCN and behavior.


Subject(s)
Circadian Clocks/genetics , Circadian Rhythm/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Motor Activity/physiology , Suprachiasmatic Nucleus/physiology , Transcription, Genetic , ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/genetics , Animals , Core Binding Factor Alpha 1 Subunit/deficiency , Gene Expression Regulation , Male , Mice , Mice, Transgenic , Olfactory Bulb/physiology , Paraventricular Hypothalamic Nucleus/physiology , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
20.
Circ Res ; 111(5): 543-52, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22773442

ABSTRACT

RATIONALE: Vascular calcification is a hallmark of atherosclerosis, a major cause of morbidity and mortality in the United States. We have previously reported that the osteogenic transcription factor Runx2 is an essential and sufficient regulator of calcification of vascular smooth muscle cells (VSMC) in vitro. OBJECTIVE: To determine the contribution of osteogenic differentiation of VSMC to the pathogenesis of vascular calcification and the function of VSMC-derived Runx2 in regulating calcification in vivo. METHODS AND RESULTS: SMC-specific Runx2-deficient mice, generated by breeding SM22α-Cre mice with the Runx2 exon 8 floxed mice, exhibited normal aortic gross anatomy and expression levels of SMC-specific marker genes. Runx2 deficiency did not affect basal SMC markers, but inhibited oxidative stress-reduced expression of SMC markers. High-fat-diet-induced vascular calcification in vivo was markedly inhibited in the Runx2-deficient mice in comparison with their control littermates. Runx2 deficiency inhibited the expression of receptor activator of nuclear factor κB ligand, which was accompanied by decreased macrophage infiltration and formation of osteoclast-like cells in the calcified lesions. Coculture of VSMC with bone marrow-derived macrophages demonstrated that the Runx2-deficient VSMC failed to promote differentiation of macrophages into osteoclast-like cells. CONCLUSIONS: These data have determined the importance of osteogenic differentiation of VSMC in the pathogenesis of vascular calcification in mice and defined the functional role of SMC-derived Runx2 in regulating vascular calcification and promoting infiltration of macrophages into the calcified lesion to form osteoclast-like cells. Our studies suggest that the development of vascular calcification is coupled with the formation of osteoclast-like cells, paralleling the bone remodeling process.


Subject(s)
Atherosclerosis/pathology , Calcinosis/pathology , Core Binding Factor Alpha 1 Subunit/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/physiology , Acid Phosphatase/metabolism , Animals , Atherosclerosis/physiopathology , Bone Remodeling/physiology , Calcinosis/physiopathology , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Core Binding Factor Alpha 1 Subunit/deficiency , Diet, High-Fat , Disease Models, Animal , Exons/genetics , Female , Isoenzymes/metabolism , Macrophages/cytology , Male , Mice , Mice, Knockout , Mutagenesis/physiology , Osteoclasts/cytology , RANK Ligand/genetics , RANK Ligand/metabolism , Tartrate-Resistant Acid Phosphatase
SELECTION OF CITATIONS
SEARCH DETAIL
...