1
|
Hong HS, Park HJ, Lee JM, Chen ZY, Kim TW, Seo YS, Kang JW, Seo YK. Bioactive Carbon Dots from Clove Residue: Synthesis, Characterization, and Osteogenic Properties. Biomedicines 2025; 13:527. [PMID: 40002941 PMCID: PMC11852471 DOI: 10.3390/biomedicines13020527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Bone regeneration using nanomaterial-based approaches shows promise for treating critical bone defects. However, developing sustainable and cost-effective therapeutic materials remains challenging. This study investigates the osteogenic potential of clove-derived carbon dots (C-CDs) for bone regeneration applications. Methods: C-CDs were synthesized using a green hydrothermal method. The osteogenic potential was evaluated in human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and validated using ectopic bone formation and calvarial defect models. Results: C-CDs demonstrated uniform morphology (~10 nm) with efficient cellular uptake. In vitro studies showed successful osteogenic differentiation through the upregulation of RUNX2, ALP, COL1A1, and BMP-2 mediated by Wnt/β-catenin/GSK3β and BMP signaling pathways. In vivo models have also demonstrated that C-CDs are effective in promoting bone regeneration. Conclusions: These findings establish C-CDs as promising candidates for bone regeneration therapy, offering a sustainable alternative to current treatments. While optimization is needed, their demonstrated osteogenic properties warrant further development for regenerative medicine applications.
Collapse
Affiliation(s)
- Hye-Sun Hong
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| | - Hee-Jung Park
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| | - Ji-Min Lee
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| | - Zu-Yu Chen
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| | - Tae-Woo Kim
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| | - Yong-Seok Seo
- Department of Food Science and Biotechnology, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea;
| | - Jun-Won Kang
- Department of Food Science and Biotechnology, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea;
| | - Young-Kwon Seo
- Department of Biomedical Engineering, Dongguk University, Goyang-si 10326, Gyeonggi-do, Republic of Korea; (H.-S.H.); (H.-J.P.); (J.-M.L.); (Z.-Y.C.); (T.-W.K.)
| |
Collapse
|
2
|
Rahaman SH, Bodhak S, Balla VK, Bhattacharya D. Role of in-situ electrical stimulation on early-stage mineralization and in-vitro osteogenesis of electroactive bioactive glass composites. BIOMATERIALS ADVANCES 2025; 166:214062. [PMID: 39406157 DOI: 10.1016/j.bioadv.2024.214062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/07/2024] [Indexed: 11/13/2024]
Abstract
Bioactive glass (BAG) has emerged as an effective bone graft substitute due to its diverse qualities of biocompatibility, bioactivity, osteoblast adhesion and enhanced revascularization. However, inferior osteogenic capacity of BAG compared to autologous bone grafts continues to limiting it's wide-spread clinical applications towards repairing of bone fractures and healing. In this study, we have fabricated BAG composites with 0.5 to 2 wt% bismuth ferrite (BF, a multiferroic material) with an aim to generate in-situ electrical charges pertinent to early-stage bone regeneration thus mimicking natural bone, which is a piezoelectric material. The fabricated BAG composites were characterised in terms of microstructures, phase analysis, remanent polarization, wettability and subsequently evaluated for in vitro cell proliferation and osteogenesis with and without magnetic field exposure (200 mT, 30 min./day). Pre-osteoblast cells from mice (MC3T3-E1) seeded on these composites exhibited excellent cell growth without any cytotoxicity, which is further supported by FITC/DAPI staining and a live/dead assay. The results of Alizarin Red S assay and increased levels of Alkaline Phosphatase (ALP) activity, at 21 days of culture, suggest that the BAG-BF composites promote in vitro osteogenic differentiation of pre-osteoblast cells. The enhanced osteogenesis of BAG-BF composites was also confirmed through qRT-PCR analysis, which showed rapid upregulation of osteoblastogenic specific genes namely RunX-2, Collagen-1, Bone Sialo Protein, and ALP after 21 days. Additionally, the osteogenic differentiation was assessed by the Western Blot technique, which revealed significantly higher band intensity of osteogenic markers in BAG-1.5 BF and BAG-2 BF composites than pure BAG. These findings clearly demonstrate that in-situ electrical stimulation and osteoconductive capacity of BF reinforced BAG composites have positive impact on osteoblast cell development, bone formation, and healing.
Collapse
Affiliation(s)
- Sk Hasanur Rahaman
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subhadip Bodhak
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Vamsi Krishna Balla
- Bioceramics and Coating Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Dipten Bhattacharya
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; Advanced Mechanical and Materials Characterization Division, CSIR-Central Glass and Ceramic Research Institute, 196, Raja S.C Mullick Road, Jadavpur, Kolkata 700 032, India
| |
Collapse
|
3
|
Boanini E, Pagani S, Gazzano M, Rubini K, Raimondi L, De Luca A, Romanelli A, Giavaresi G, Bigi A. Mn 2+ vs Co 2+ substitution into β-TCP: Structural details and bone cells response. Colloids Surf B Biointerfaces 2024; 243:114154. [PMID: 39137528 DOI: 10.1016/j.colsurfb.2024.114154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/24/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
This work investigated the range of substitution of two biologically relevant ions, namely Mn2+ and Co2+, into the structure of β-tricalcium phosphate, as well as their influence on bone cells response. To this aim, β-TCP was synthesized by solid state reaction in the presence of increasing amount of the substituent ions. The results of the X-ray diffraction analysis reveal that just limited amounts of these ions can enter into the β-TCP structure: 15 at% and 20 at% for cobalt and manganese, respectively. Substitution provokes aggregation of the micrometric particles and reduction of the lattice constants. In particular, the dimension of the c-parameter exhibits a discontinuity at about 10 at% for both cations, although with different trend. Moreover, Rietveld refinement demonstrates a clear preference of both manganese and cobalt for the octahedral site (V). The influence of these ions on cell response was tested on osteoblast, osteoclast and endothelial cells. The results indicate that the presence of manganese promotes a good osteoblast viability, significantly enhances the expression of osteoblast key genes and the angiogenic process of endothelial cells, while inhibiting osteoclast resorption. At variance, osteoblast viability appears reduced in the presence of Co samples, on which osteoblast genes reach higher expression than on β-TCP just in a few cases. On the other hand, the results clearly show that cobalt significantly stimulates the angiogenic process and inhibits osteoclast resorption.
Collapse
Affiliation(s)
- Elisa Boanini
- Department of Chemistry ''Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy.
| | - Stefania Pagani
- CS-Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | | | - Katia Rubini
- Department of Chemistry ''Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| | - Lavinia Raimondi
- CS-Surgical Sciences and Technologies-SS Omics Science Platform for Personalized Orthopedics, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Angela De Luca
- CS-Surgical Sciences and Technologies-SS Omics Science Platform for Personalized Orthopedics, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Alessia Romanelli
- CS-Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Gianluca Giavaresi
- CS-Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, Bologna 40136, Italy
| | - Adriana Bigi
- Department of Chemistry ''Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, Bologna 40126, Italy
| |
Collapse
|
4
|
Silingardi F, Salamanna F, Español M, Maglio M, Sartori M, Giavaresi G, Bigi A, Ginebra MP, Boanini E. Regulation of osteogenesis and angiogenesis by cobalt, manganese and strontium doped apatitic materials for functional bone tissue regeneration. BIOMATERIALS ADVANCES 2024; 163:213968. [PMID: 39059113 DOI: 10.1016/j.bioadv.2024.213968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 05/13/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Strontium, cobalt, and manganese ions are present in the composition of bone and useful for bone metabolism, even when combined with calcium phosphate in the composition of biomaterials. Herein we explored the possibility to include these ions in the composition of apatitic materials prepared through the cementitious reaction between ion-substituted calcium phosphate dibasic dihydrate, CaHPO4·2H2O (DCPD) and tetracalcium phosphate, Ca4(PO4)2O (TTCP). The results of the chemical, structural, morphological and mechanical characterization indicate that cobalt and manganese exhibit a greater delaying effect than strontium (about 15 at.%) on the cementitious reaction, even though they are present in smaller amounts within the materials (about 0.8 and 4.5 at.%, respectively). Furthermore, the presence of the foreign ions in the apatitic materials leads to a slight reduction of porosity and to enhancement of compressive strength. The results of biological tests show that the presence of strontium and manganese, as well as calcium, in the apatitic materials cultured in direct contact with human mesenchymal stem cells (hMSCs) stimulates their viability and activity. In contrast, the apatitic material containing cobalt exhibits a lower metabolic activity. All the materials have a positive effect on the expression of Vascular Endothelial Growth Factor (VEGF) and Von Willebrand Factor (vWF). Moreover, the apatitic material containing strontium induces the most significant reduction in the differentiation of preosteoclasts into osteoclasts, demonstrating not only osteogenic and angiogenic properties, but also ability to regulate bone resorption.
Collapse
Affiliation(s)
- Francesca Silingardi
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Francesca Salamanna
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Montserrat Español
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona 08019, Spain
| | - Melania Maglio
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Maria Sartori
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Gianluca Giavaresi
- Surgical Sciences and Technologies, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Adriana Bigi
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy
| | - Maria-Pau Ginebra
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering, Universitat Politècnica de Catalunya (UPC), Barcelona 08019, Spain
| | - Elisa Boanini
- Department of Chemistry "Giacomo Ciamician", Alma Mater Studiorum - University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
5
|
Vuong TA, Zhang Y, Kim J, Leem YE, Kang JS. Prmt7 is required for the osteogenic differentiation of mesenchymal stem cells via modulation of BMP signaling. BMB Rep 2024; 57:330-335. [PMID: 38627951 PMCID: PMC11289507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/13/2023] [Accepted: 01/15/2024] [Indexed: 08/03/2024] Open
Abstract
Arginine methylation, which is catalyzed by protein arginine methyltransferases (Prmts), is known to play a key role in various biological processes. However, the function of Prmts in osteogenic differentiation of mesenchymal stem cells (MSCs) has not been clearly understood. In the current study, we attempted to elucidate a positive role of Prmt7 in osteogenic differentiation. Prmt7-depleted C3H/10T1/2 cells or bone marrow mesenchymal stem cells (BMSCs) showed the attenuated expression of osteogenic specific genes and Alizarin red staining compared to the wild-type cells. Furthermore, we found that Prmt7 deficiency reduced the activation of bone morphogenetic protein (BMP) signaling cascade, which is essential for the regulation of cell fate commitment and osteogenesis. Taken together, our data indicate that Prmt7 plays important regulatory roles in osteogenic differentiation. [BMB Reports 2024; 57(7): 330-335].
Collapse
Affiliation(s)
- Tuan Anh Vuong
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Yan Zhang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - June Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Young-Eun Leem
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| |
Collapse
|
6
|
Chen Y, Liu J, Zhang Q, Chai L, Chen H, Li D, Wang Y, Qiu Y, Shen N, Zhang J, Wang Q, Wang J, Xie X, Li S, Li M. Activation of CaMKII/HDAC4 by SDF1 contributes to pulmonary arterial hypertension via stabilization Runx2. Eur J Pharmacol 2024; 970:176483. [PMID: 38479721 DOI: 10.1016/j.ejphar.2024.176483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 04/02/2024]
Abstract
Stromal derived factor 1 (SDF1) has been shown to be involved in the pathogenesis of pulmonary artery hypertension (PAH). However, the detailed molecular mechanisms remain unclear. To address this, we utilized primary cultured rat pulmonary artery smooth muscle cells (PASMCs) and monocrotaline (MCT)-induced PAH rat models to investigate the mechanisms of SDF1 driving PASMCs proliferation and pulmonary arterial remodeling. SDF1 increased runt-related transcription factor 2 (Runx2) acetylation by Calmodulin (CaM)-dependent protein kinase II (CaMKII)-dependent HDAC4 cytoplasmic translocation, elevation of Runx2 acetylation conferred its resistance to proteasome-mediated degradation. The accumulation of Runx2 further upregulated osteopontin (OPN) expression, finally leading to PASMCs proliferation. Blocking SDF1, suppression of CaMKII, inhibition the nuclear export of HDAC4 or silencing Runx2 attenuated pulmonary arterial remodeling and prevented PAH development in MCT-induced PAH rat models. Our study provides novel sights for SDF1 induction of PASMCs proliferation and suggests that targeting SDF1/CaMKII/HDAC4/Runx2 axis has potential value in the management of PAH.
Collapse
Affiliation(s)
- Yuqian Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jin Liu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Huan Chen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Danyang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Yuanjie Qiu
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Nirui Shen
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jia Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, No. 277, West Yanta Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
7
|
Upadhyay V, Singh AK, Sharma S, Sethi A, Srivastava S, Chowdhury S, Siddiqui S, Chattopadhyay N, Trivedi AK. RING finger E3 ligase, RNF138 inhibits osteoblast differentiation by negatively regulating Runx2 protein turnover. J Cell Physiol 2024; 239:e31217. [PMID: 38327035 DOI: 10.1002/jcp.31217] [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: 08/28/2023] [Revised: 01/23/2024] [Accepted: 01/27/2024] [Indexed: 02/09/2024]
Abstract
A few ubiquitin ligases have been shown to target Runx2, the key osteogenic transcription factor and thereby regulate bone formation. The regulation of Runx2 expression and function are controlled both at the transcriptional and posttranslational levels. Really interesting new gene (RING) finger ubiquitin ligases of which RNF138 is a member are important players in the ubiquitin-proteasome system, contributing to the regulation of protein turnover and cellular processes. Here, we demonstrated that RNF138 negatively correlated with Runx2 protein levels in osteopenic ovariectomized rats which implied its role in bone loss. Accordingly, RNF138 overexpression potently inhibited osteoblast differentiation of mesenchyme-like C3H10T1/2 as well primary rat calvarial osteoblast (RCO) cells in vitro, whereas overexpression of catalytically inactive mutant RNF138Δ18-58 (lacks RING finger domain) had mild to no effect. Contrarily, RNF138 depletion copiously enhanced endogenous Runx2 levels and augmented osteogenic differentiation of C3H10T1/2 as well as RCOs. Mechanistically, RNF138 physically associates within multiple regions of Runx2 and ubiquitinates it leading to its reduced protein stability in a proteasome-dependent manner. Moreover, catalytically active RNF138 destabilized Runx2 which resulted in inhibition of its transactivation potential and physiological function of promoting osteoblast differentiation leading to bone loss. These findings underscore the functional involvement of RNF138 in bone formation which is primarily achieved through its modulation of Runx2 by stimulating ubiquitin-mediated proteasomal degradation. Thus, our findings indicate that RNF138 could be a promising novel target for therapeutic intervention in postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Vishal Upadhyay
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anil Kumar Singh
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shivani Sharma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
| | - Arppita Sethi
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Swati Srivastava
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
| | - Sangita Chowdhury
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shumaila Siddiqui
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Naibedya Chattopadhyay
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
| | - Arun Kumar Trivedi
- Division of Cancer Biology, CSIR-Central Drug Research Institute, Lucknow, Utter Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
8
|
Shiv R, Rakeswari, Farjana N, Subbiah U, Ajith A, Balaji A, Mohanasatheesh S. Characterization of missense nonsynonymous single-nucleotide polymorphism of runt-related transcription factor-2 gene - An in silico approach. Indian J Pharmacol 2024; 56:198-205. [PMID: 39078184 PMCID: PMC11286098 DOI: 10.4103/ijp.ijp_533_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/23/2024] [Accepted: 06/04/2024] [Indexed: 07/31/2024] Open
Abstract
OBJECTIVES Single-nucleotide polymorphism (SNP) codes for multiple amino acids, impacting protein functions and disease prognosis. Runt-related transcription factor-2 (RUNX2), a transcription factor linked to osteoblast differentiation, regulates cell proliferation in endothelium and osteoblastic cells. Understanding Runx2's role in nonosseous tissues is rapidly advancing. This study aims to identify harmful SNPs of the RUNX2 gene that may alter disease susceptibility using computational techniques. METHODS The study uses various in silico methods to identify nonsynonymous SNPs (nsSNPs) of the RUNX2 gene, which could potentially alter protein structure and functions, with further analyses by I-Mutant, ConSurf, Netsurf 3.0, GeneMANIA, and Have (y)Our Protein Explained. RESULTS Six missense nsSNPs were identified as potentially harmful, disease-causing, and damaging. Four were found to be unstable, while five were conserved. All six nsSNPs had a coiled secondary structure. Five nsSNPs were found to be destabilized. CONCLUSION The RUNX2 gene's deleterious missense nsSNPs were identified by this study, and they may be exploited in future experimental studies. These high-risk nsSNPs might be considered target molecules in therapeutic and diagnostic therapies in teeth and bone development.
Collapse
Affiliation(s)
- Ragul Shiv
- Department of Periodontics, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Rakeswari
- Department of Periodontics, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Nilofer Farjana
- Department of Periodontics, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Usha Subbiah
- Human Genetics Research Centre, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Athira Ajith
- Human Genetics Research Centre, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Anitha Balaji
- Department of Periodontics, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - S. Mohanasatheesh
- Department of Periodontics, Sree Balaji Dental College and Hospital, Bharath Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| |
Collapse
|
9
|
Liu X, Song J, Yan X, Li P, Zhang J, Wang B, Si J, Chen Y. N-nitrosodimethylamine exposure to zebrafish embryos/larvae causes cardiac and spinal developmental toxicity. Comp Biochem Physiol C Toxicol Pharmacol 2024; 277:109823. [PMID: 38158031 DOI: 10.1016/j.cbpc.2023.109823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/27/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
N-nitrosodimethylamine (NDMA), one of the new nitrogen-containing disinfection by-products, is potentially cytotoxic, genotoxic, and carcinogenic. Its potential toxicological effects have attracted a wide range of attention, but the mechanism is still not sufficiently understood. To better understand the toxicological mechanisms of NDMA, zebrafish embryos were exposed to NDMA from 3 h post-fertilization (hpf) to 120hpf. Mortality and malformation were significantly increased, and hatching rate, heart rate, and swimming behavior were decreased in the exposure groups. The result indicated that NDMA exposure causes cardiac and spinal developmental toxicity. mRNA levels of genes involved in the apoptotic pathway, including p53, bax, and bcl-2 were significantly affected by NDMA exposure. Moreover, the genes associated with spinal and cardiac development (myh6, myh7, nkx2.5, eph, bmp2b, bmp4, bmp9, run2a, and run2b) were significantly downregulated after treatment with NDMA. Wnt and TGF-β signaling pathways, crucial for the development of diverse tissues and organs in the embryo and the establishment of the larval spine, were also significantly disturbed by NDMA treatment. In summary, the disinfection by-product, NDMA, exhibits spinal and cardiac developmental toxicity in zebrafish embryos, providing helpful information for comprehensive analyses and a better understanding the mechanism of its toxicity.
Collapse
Affiliation(s)
- Xiaoyi Liu
- College of Life Science, Lanzhou University, Lanzhou, China. https://twitter.com/@LanoLiu41230
| | - Jinge Song
- School of Stomatology, Lanzhou University, Lanzhou, China
| | - Xiaotao Yan
- Lanzhou Urban Water Supply (Group) Co., Ltd, Lanzhou, China
| | - Pingping Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jinhua Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Bin Wang
- Lanzhou Urban Water Supply (Group) Co., Ltd, Lanzhou, China
| | - Jing Si
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
| | - Yong Chen
- College of Life Science, Lanzhou University, Lanzhou, China.
| |
Collapse
|
10
|
Tang K, Sun L, Chen L, Feng X, Wu J, Guo H, Zheng Y. Bioinformatics Analysis and Experimental Validation of Mitochondrial Autophagy Genes in Knee Osteoarthritis. Int J Gen Med 2024; 17:639-650. [PMID: 38414629 PMCID: PMC10898481 DOI: 10.2147/ijgm.s444847] [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: 10/15/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Background Mitochondrial autophagy is closely related to the pathogenesis of osteoarthritis, In order to explore the role of mitochondrial autophagy related genes in knee osteoarthritis (KOA) and its molecular mechanism. Methods KOA-related transcriptome data were extracted from the Gene Expression Omnibus (GEO) database. Differentially expressed mitochondrial autophagy gene (DEMGs) were screened in patients with KOA by differential expression analysis. The STRING website was used to construct a protein-protein interaction (PPI) network among DEMGs. Molecular complex detection (MCODE) method in Cytoscape software was performed to identify hub DEMGs. Support vector machine recursive feature elimination (SVM-RFE) method was used to construct the hub DEMG diagnosis model. Genes with diagnostic value were identified as biomarkers by plotting receiver operating characteristic (ROC) curves and Expression validation. CIBERSORT algorithm was used to calculate the proportion of 22 immune cells in each sample in the GSE114007 dataset. Finally, biomarker expression was verified by qPCR. Results A total of 15 DEMGs were obtained and enrichment analyses showed that these DEMG strains were mainly enriched in the mitophagy-animal, shigellosis, autophagy-animal and FoxO signal pathways. The PPI network unveiled 13 DEMGs with interactions. In addition, 8 hub DEMGs (ULK1, CALCOCO2, MAP1LC3B, BNIP3L, GABARAPL1, BNIP3, FKBP8 and FOXO3) were obtained for KOA. And 5 model DEMGs (BNIP3L, BNIP3, MAP1LC3B, ULK1 and FOXO3) were screened. The ROC curves revealed that BNIP3 and FOXO3 has strong diagnostic value in these models of DEMG. Immune-infiltration and correlation analysis showed that BNIP3 and FOXO3 were significantly correlated with three different immune cells, including primary B cells, M0 macrophage and M2 macrophage. The cartilage tissue samples qPCR verification results show that FOXO3 and BNIP3 were all down-regulated in KOA (p < 0.01), and the validation results are consistent with the above analysis. Conclusion BNIP3 and FOXO3 have been identified as biomarkers for the diagnosis of KOA, which might supply a new insight for the pathogenesis and treatment of KOA.
Collapse
Affiliation(s)
- Kuihan Tang
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| | - Li Sun
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Long Chen
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Xiaobo Feng
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| | - Jiarui Wu
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Hao Guo
- Department of Orthopedics, Guizhou Provincial People’s Hospital, Guiyang, 550000, People’s Republic of China
| | - Yong Zheng
- Department of Orthopedics, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, 550014, People’s Republic of China
| |
Collapse
|
11
|
Biguetti CC, Arteaga A, Chandrashekar BL, Rios E, Margolis R, Rodrigues DC. A Model of Immediate Implant Placement to Evaluate Early Osseointegration in 129/Sv Diabetic Mice. Int J Oral Maxillofac Implants 2023; 38:1200-1210. [PMID: 38085752 PMCID: PMC11181517 DOI: 10.11607/jomi.10335] [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] [Indexed: 12/18/2023] Open
Abstract
PURPOSE To analyze the process of early oral osseointegration of titanium (Ti) implants in diabetic 129/Sv mice through microCT and histologic and immunohistochemical analysis. MATERIALS AND METHODS A group of 30 male 129/Sv mice was equally subdivided into two groups: (1) nondiabetic (ND), in which mice did not undergo systemic alterations and received a standard diet, and (2) diabetic (D), in which mice were provided a high-fat diet from the age of 6 weeks until the conclusion of the study and received two intraperitoneal (IP) injections of streptozotocin (STZ) at a concentration of 100 mg/Kg each. Each mouse underwent extraction of a maxillary first molar, and customized Ti screws (0.50 mm diameter, 1.5 mm length) were placed in the residual alveolar sockets of the palatal roots. At 7 and 21 days after implant placement, the animals were euthanized for maxilla and pancreas collection. Maxillae containing Ti implants were analyzed with microCT, histology, and immunohistochemistry for cells that were positive for F4/80, CD146, runt-related transcription factor 2 (Runx2), and proliferating cell nuclear antigen (PCNA). Pancreata were histologically analyzed. Quantitative data were statistically analyzed with a significance level at 5% (P < .05). RESULTS ND mice presented successful healing and osseointegration, with a significantly higher fraction of bone volume compared to D mice, both at the alveolar sockets (53.39 ± 5.93 and 46.08 ± 3.18, respectively) and at the implant sites (68.88 ± 7.07 and 44.40 ± 6.98, respectively) 21 days after implant placement. Histologic evaluation revealed that the ND mice showed a significant decrease in inflammatory infiltrate and a significant increase in newly formed bone matrix at 21 days, whereas peri-implant sites in the D mice were predominantly encapsulated by fibrous tissue and chronic inflammatory infiltrate. Immunohistochemical characterization revealed higher Runx2 osteoblast differentiation and higher cell proliferation activity in the ND mice at 7 days, while higher amounts of macrophages were present in D mice at 7 and 21 days. Interestingly, no differences were found in CD146-positive cells when comparing ND and D mice. CONCLUSIONS This study evaluated the effects of immediate dental implant placement in 129/Sv diabetic mice by using specific healing markers to identify changes in cellular events involved in early oral osseointegration. This approach may serve as tool to evaluate new materials and surface coatings to improve osseointegration in diabetic patients.
Collapse
|
12
|
Khotib J, Marhaeny HD, Miatmoko A, Budiatin AS, Ardianto C, Rahmadi M, Pratama YA, Tahir M. Differentiation of osteoblasts: the links between essential transcription factors. J Biomol Struct Dyn 2023; 41:10257-10276. [PMID: 36420663 DOI: 10.1080/07391102.2022.2148749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/12/2022] [Indexed: 11/27/2022]
Abstract
Osteoblasts, cells derived from mesenchymal stem cells (MSCs) in the bone marrow, are cells responsible for bone formation and remodeling. The differentiation of osteoblasts from MSCs is triggered by the expression of specific genes, which are subsequently controlled by pro-osteogenic pathways. Mature osteoblasts then differentiate into osteocytes and are embedded in the bone matrix. Dysregulation of osteoblast function can cause inadequate bone formation, which leads to the development of bone disease. Various key molecules are involved in the regulation of osteoblastogenesis, which are transcription factors. Previous studies have heavily examined the role of factors that control gene expression during osteoblastogenesis, both in vitro and in vivo. However, the systematic relationship of these transcription factors remains unknown. The involvement of ncRNAs in this mechanism, particularly miRNAs, lncRNAs, and circRNAs, has been shown to influence transcriptional factor activity in the regulation of osteoblast differentiation. Here, we discuss nine essential transcription factors involved in osteoblast differentiation, including Runx2, Osx, Dlx5, β-catenin, ATF4, Ihh, Satb2, and Shn3. In addition, we summarize the role of ncRNAs and their relationship to these essential transcription factors in order to improve our understanding of the transcriptional regulation of osteoblast differentiation. Adequate exploration and understanding of the molecular mechanisms of osteoblastogenesis can be a critical strategy in the development of therapies for bone-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Honey Dzikri Marhaeny
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aniek Setiya Budiatin
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Mahardian Rahmadi
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Yusuf Alif Pratama
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Tahir
- Department of Pharmaceutical Science, Kulliyah of Pharmacy, International Islamic University Malaysia, Pahang, Malaysia
| |
Collapse
|
13
|
Piao M, Lee SH, Li Y, Choi JK, Yeo CY, Lee KY. Cyclophilin E (CypE) Functions as a Positive Regulator in Osteoblast Differentiation by Regulating the Transcriptional Activity of Runx2. Cells 2023; 12:2549. [PMID: 37947627 PMCID: PMC10648996 DOI: 10.3390/cells12212549] [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: 10/11/2023] [Revised: 10/20/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Cyclophilin E (CypE) belongs to the cyclophilin family and exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity. It participates in various biological processes through the regulation of peptidyl-prolyl isomerization. However, the specific role of CypE in osteoblast differentiation has not yet been elucidated. In this study, we first discovered the positive impact of CypE on osteoblast differentiation through gain or loss of function experiments. Mechanistically, CypE enhances the transcriptional activity of Runx2 through its PPIase activity. Furthermore, we identified the involvement of the Akt signaling pathway in CypE's function in osteoblast differentiation. Taken together, our findings indicate that CypE plays an important role in osteoblast differentiation as a positive regulator by increasing the transcriptional activity of Runx2.
Collapse
Affiliation(s)
- Meiyu Piao
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Sung Ho Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Yuankuan Li
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| | - Joong-Kook Choi
- Division of Biochemistry, College of Medicine, Chungbuk National University, Cheong-Ju 28644, Republic of Korea;
| | - Chang-Yeol Yeo
- Department of Life Science and Research Center for Cellular Homeostasis, Ewha Woman’s University, Seoul 03760, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju 61186, Republic of Korea; (M.P.); (S.H.L.); (Y.L.)
| |
Collapse
|
14
|
Krishnamoorthy HS, Kannan B, Ganapathy D, Jayaseelan VP, Arumugam P. Decreased expression of the m6A RNA methyltransferase METTL3 is associated with residual ridge resorption. J Oral Biol Craniofac Res 2023; 13:563-566. [PMID: 37559689 PMCID: PMC10407425 DOI: 10.1016/j.jobcr.2023.07.003] [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: 03/10/2023] [Revised: 06/18/2023] [Accepted: 07/20/2023] [Indexed: 08/11/2023] Open
Abstract
Objective N6-methyladenosine (m6A) methylation and its regulators play crucial roles in the progression of osteoporosis (OP) by regulating the expression of osteoporosis-related genes. In this study, we have analyzed the expression of methyltransferase-like 3 (METTL3) and its target gene Runt-related transcription factor 2 (RUNX2) in patients with residual ridge resorption (RRR). Materials and methods A total 50 number of participants were included in this comparative study (RRR - n25 and healthy control - n25). Total RNA was extracted from peripheral blood and converted into cDNA. METTL3 and RUNX2 expression levels were quantified using RT-qPCR with GAPDH as the reference gene. Bioinformatics tools were used to identify gene functions and pathways. Results Real-time polymerase chain reaction (qPCR) revealed that METTL3 and RUNX2 expression was downregulated in the RRR group compared to that in healthy controls (P < 0.05). In silico functional analysis provided information regarding the role of METTL3 in various biological processes. Conclusion Our findings suggest that METTL3 dysregulation contributes to RRR pathogenesis. Further large-scale samples and functional studies are required to identify their therapeutic potential.
Collapse
Affiliation(s)
- Harini Sri Krishnamoorthy
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Balachander Kannan
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Vijayashree Priyadharsini Jayaseelan
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Paramasivam Arumugam
- Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| |
Collapse
|
15
|
Li X, Zhang J, Wang B, Chen C, Zhang E, Lv Z, He Q, Hu Y, Wang X, Zhang F. USP24-dependent stabilization of Runx2 recruits a p300/NCOA3 complex to transactivate ADAMTS genes and promote degeneration of intervertebral disc in chronic inflammation mice. Biol Direct 2023; 18:37. [PMID: 37415159 PMCID: PMC10324278 DOI: 10.1186/s13062-023-00395-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/02/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) naturally occurs during the aging process. Its occurrence is closely related to chronic inflammation; however, the causal relationship between them is controversial. This study aimed to investigate if inflammation would promote IDD incidence and explore the underlying mechanism. METHODS A chronic inflammation mouse model was established by intraperitoneal injection of lipopolysaccharide (LPS). Enzyme-linked immunosorbent assay was performed to determine proinflammatory cytokines in serum. Histological staining was used to evaluate the degeneration of IVDs. Immunoblots and RT-qPCR analyses were performed to measure protein and mRNA expression levels. Immunoprecipitation, mass spectrometry, and co-immunoprecipitation assays were used to determine the assembly of protein complex. RESULTS We found that an inflammatory microenvironment activated p38 kinase, which phosphorylated the Runx2 transcription factor at the Ser28 site. The phosphorylated Runx2 (pRunx2) then recruited a deubiquitinase, ubiquitin-specific peptidase 24 (USP24), which stabilized pRunx2 and protected it from ubiquitin-dependent proteasomal degradation. The stabilized pRunx2 recruited histone acetyltransferase p300 and nuclear receptor coactivator 3 (NCOA3) to assemble a complex. This NCOA3-p300-pRunx2 complex then transactivated the expression of 13 ADAMTS (a disintegrin and metalloproteinase with thrombospondin motif) genes, thereby promoting the degradation of extracellular matrix (ECM) in intervertebral discs (IVDs) and causing IDD. Administration of either a p38 inhibitor (doramapimod), a NCOA3 inhibitor (bufalin), or a p300 inhibitor (EML425) significantly decreased the expression of the 13 ADAMTS genes and slowed the degeneration of IVDs. CONCLUSION In summary, our results demonstrate that USP24 protects pRunx2 from proteasomal degradation under chronic inflammation conditions, enabling pRunx2 to transactivate ADAMTS genes and degrade ECM. Our findings provide direct evidence that chronic inflammation triggers IDD and offer a therapeutic strategy for retarding IDD in patients with chronic inflammation.
Collapse
Affiliation(s)
- Xingguo Li
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Jun Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Bing Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Chao Chen
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Enyu Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Zhengpin Lv
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Qicong He
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yaoquan Hu
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Xuenan Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Fan Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China.
| |
Collapse
|
16
|
Zhang Q, Li Q, Wang Y, Zhang Y, Peng R, Wang Z, Zhu B, Xu L, Gao X, Chen Y, Gao H, Hu J, Qian C, Ma M, Duan R, Li J, Zhang L. Characterization of Chromatin Accessibility in Fetal Bovine Chondrocytes. Animals (Basel) 2023; 13:1875. [PMID: 37889831 PMCID: PMC10251841 DOI: 10.3390/ani13111875] [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: 04/14/2023] [Revised: 05/28/2023] [Accepted: 05/29/2023] [Indexed: 10/29/2023] Open
Abstract
Despite significant advances of the bovine epigenome investigation, new evidence for the epigenetic basis of fetal cartilage development remains lacking. In this study, the chondrocytes were isolated from long bone tissues of bovine fetuses at 90 days. The Assay for Transposase-Accessible Chromatin with high throughput sequencing (ATAC-seq) and transcriptome sequencing (RNA-seq) were used to characterize gene expression and chromatin accessibility profile in bovine chondrocytes. A total of 9686 open chromatin regions in bovine fetal chondrocytes were identified and 45% of the peaks were enriched in the promoter regions. Then, all peaks were annotated to the nearest gene for Gene Ontology (GO) and Kyoto Encylopaedia of Genes and Genomes (KEGG) analysis. Growth and development-related processes such as amide biosynthesis process (GO: 0043604) and translation regulation (GO: 006417) were enriched in the GO analysis. The KEGG analysis enriched endoplasmic reticulum protein processing signal pathway, TGF-β signaling pathway and cell cycle pathway, which are closely related to protein synthesis and processing during cell proliferation. Active transcription factors (TFs) were enriched by ATAC-seq, and were fully verified with gene expression levels obtained by RNA-seq. Among the top50 TFs from footprint analysis, known or potential cartilage development-related transcription factors FOS, FOSL2 and NFY were found. Overall, our data provide a theoretical basis for further determining the regulatory mechanism of cartilage development in bovine.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Qian Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yahui Wang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yapeng Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Ruiqi Peng
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Zezhao Wang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Bo Zhu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Lingyang Xu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Xue Gao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Yan Chen
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Huijiang Gao
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Junwei Hu
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Cong Qian
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Minghao Ma
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Rui Duan
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| | - Junya Li
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
| | - Lupei Zhang
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Q.Z.); (Q.L.); (Y.W.); (Y.Z.); (R.P.); (Z.W.); (B.Z.); (L.X.); (X.G.); (Y.C.); (H.G.)
- Academy of Pingliang Red Cattle, 492 South Ring Road, Kongtong District, Pingliang 744000, China; (J.H.); (C.Q.); (M.M.); (R.D.)
| |
Collapse
|
17
|
Man K, Eisenstein NM, Hoey DA, Cox SC. Bioengineering extracellular vesicles: smart nanomaterials for bone regeneration. J Nanobiotechnology 2023; 21:137. [PMID: 37106449 PMCID: PMC10134574 DOI: 10.1186/s12951-023-01895-2] [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: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as key regulators of bone development, homeostasis and repair. EV-based therapies have the potential to circumnavigate key issues hindering the translation of cell-based therapies including functional tissue engraftment, uncontrolled differentiation and immunogenicity issues. Due to EVs' innate biocompatibility, low immunogenicity, and high physiochemical stability, these naturally-derived nanoparticles have garnered growing interest as potential acellular nanoscale therapeutics for a variety of diseases. Our increasing knowledge of the roles these cell-derived nanoparticles play, has made them an exciting focus in the development of novel pro-regenerative therapies for bone repair. Although these nano-sized vesicles have shown promise, their clinical translation is hindered due to several challenges in the EV supply chain, ultimately impacting therapeutic efficacy and yield. From the biochemical and biophysical stimulation of parental cells to the transition to scalable manufacture or maximising vesicles therapeutic response in vivo, a multitude of techniques have been employed to improve the clinical efficacy of EVs. This review explores state of the art bioengineering strategies to promote the therapeutic utility of vesicles beyond their native capacity, thus maximising the clinical potential of these pro-regenerative nanoscale therapeutics for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Neil M Eisenstein
- Research and Clinical Innovation, Royal Centre for Defence Medicine, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, UK
- Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham, B15 2TH, UK
| | - David A Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
- Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College, Dublin 2, D02 DK07, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, D02 VN51, Dublin, Ireland
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
18
|
Li X, Kaur N, Albahrani M, Karpf AR, Black AR, Black JD. Crosstalk between protein kinase C α and transforming growth factor β signaling mediated by Runx2 in intestinal epithelial cells. J Biol Chem 2023; 299:103017. [PMID: 36791912 PMCID: PMC10036670 DOI: 10.1016/j.jbc.2023.103017] [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: 05/10/2022] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/15/2023] Open
Abstract
Tight coordination of growth regulatory signaling is required for intestinal epithelial homeostasis. Protein kinase C α (PKCα) and transforming growth factor β (TGFβ) are negative regulators of proliferation with tumor suppressor properties in the intestine. Here, we identify novel crosstalk between PKCα and TGFβ signaling. RNA-Seq analysis of nontransformed intestinal crypt-like cells and colorectal cancer cells identified TGFβ receptor 1 (TGFβR1) as a target of PKCα signaling. RT-PCR and immunoblot analysis confirmed that PKCα positively regulates TGFβR1 mRNA and protein expression in these cells. Effects on TGFβR1 were dependent on Ras-extracellular signal-regulated kinase 1/2 (ERK) signaling. Nascent RNA and promoter-reporter analysis indicated that PKCα induces TGFβR1 transcription, and Runx2 was identified as an essential mediator of the effect. PKCα promoted ERK-mediated activating phosphorylation of Runx2, which preceded transcriptional activation of the TGFβR1 gene and induction of Runx2 expression. Thus, we have identified a novel PKCα→ERK→Runx2→TGFβR1 signaling axis. In further support of a link between PKCα and TGFβ signaling, PKCα knockdown reduced the ability of TGFβ to induce SMAD2 phosphorylation and cell cycle arrest, and inhibition of TGFβR1 decreased PKCα-induced upregulation of p21Cip1 and p27Kip1 in intestinal cells. The physiological relevance of these findings is also supported by The Cancer Genome Atlas data showing correlation between PKCα, Runx2, and TGFβR1 mRNA expression in human colorectal cancer. PKCα also regulated TGFβR1 in endometrial cancer cells, and PKCα, Runx2, and TGFβR1 expression correlates in uterine tumors, indicating that crosstalk between PKCα and TGFβ signaling may be a common mechanism in diverse epithelial tissues.
Collapse
Affiliation(s)
- Xinyue Li
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Navneet Kaur
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Mustafa Albahrani
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Adam R Karpf
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| |
Collapse
|
19
|
Lu C, Zhao H, Liu Y, Yang Z, Yao H, Liu T, Gou T, Wang L, Zhang J, Tian Y, Yang Y, Zhang H. Novel Role of the SIRT1 in Endocrine and Metabolic Diseases. Int J Biol Sci 2023; 19:484-501. [PMID: 36632457 PMCID: PMC9830516 DOI: 10.7150/ijbs.78654] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/15/2022] [Indexed: 12/23/2022] Open
Abstract
Silent information regulator 1 (SIRT1), a highly conserved NAD+-dependent deacetylase, is a cellular regulator that has received extensive attention in recent years and regarded as a sensor of cellular energy and metabolism. The accumulated evidence suggests that SIRT1 is involved in the development of endocrine and metabolic diseases. In a variety of organisms, SIRT1 regulates gene expression through the deacetylation of histone, transcription factors, and lysine residues of other modified proteins including several metabolic and endocrine signal transcription factors, thereby enhancing the therapeutic effects of endocrine and metabolic diseases. These evidences indicate that targeting SIRT1 has promising applications in the treatment of endocrine and metabolic diseases. This review focuses on the role of SIRT1 in endocrine and metabolic diseases. First, we describe the background and structure of SIRT1. Then, we outline the role of SIRT1 in endocrine and metabolic diseases such as hyperuricemia, diabetes, hypertension, hyperlipidemia, osteoporosis, and polycystic ovarian syndrome. Subsequently, the SIRT1 agonists and inhibitors in the above diseases are summarized and future research directions are proposed. Overall, the information presents here may highlight the potential of SIRT1 as a future biomarker and therapeutic target for endocrine and metabolic diseases.
Collapse
Affiliation(s)
- Chenxi Lu
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Yanqing Liu
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Airforce Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Hairong Yao
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Tong Liu
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Tiantian Gou
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Li Wang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Juan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ye Tian
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,✉ Corresponding authors: Yang Yang: . Huan Zhang: . Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| | - Huan Zhang
- Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China.,✉ Corresponding authors: Yang Yang: . Huan Zhang: . Department of Cardiology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University. Faculty of Life Sciences and Medicine, Northwest University, 10 Fengcheng Three Road, Xi'an, China
| |
Collapse
|
20
|
Kitajima H, Hirota M, Komatsu K, Isono H, Matsuura T, Mitsudo K, Ogawa T. Ultraviolet Light Treatment of Titanium Microfiber Scaffolds Enhances Osteoblast Recruitment and Osteoconductivity in a Vertical Bone Augmentation Model: 3D UV Photofunctionalization. Cells 2022; 12:cells12010019. [PMID: 36611812 PMCID: PMC9818481 DOI: 10.3390/cells12010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Vertical bone augmentation to create host bone prior to implant placement is one of the most challenging regenerative procedures. The objective of this study is to evaluate the capacity of a UV-photofunctionalized titanium microfiber scaffold to recruit osteoblasts, generate intra-scaffold bone, and integrate with host bone in a vertical augmentation model with unidirectional, limited blood supply. Scaffolds were fabricated by molding and sintering grade 1 commercially pure titanium microfibers (20 μm diameter) and treated with UVC light (200-280 nm wavelength) emitted from a low-pressure mercury lamp for 20 min immediately before experiments. The scaffolds had an even and dense fiber network with 87% porosity and 20-50 mm inter-fiber distance. Surface carbon reduced from 30% on untreated scaffold to 10% after UV treatment, which corresponded to hydro-repellent to superhydrophilic conversion. Vertical infiltration testing revealed that UV-treated scaffolds absorbed 4-, 14-, and 15-times more blood, water, and glycerol than untreated scaffolds, respectively. In vitro, four-times more osteoblasts attached to UV-treated scaffolds than untreated scaffolds three hours after seeding. On day 2, there were 70% more osteoblasts on UV-treated scaffolds. Fluorescent microscopy visualized confluent osteoblasts on UV-treated microfibers two days after seeding but sparse and separated cells on untreated microfibers. Alkaline phosphatase activity and osteocalcin gene expression were significantly greater in osteoblasts grown on UV-treated microfiber scaffolds. In an in vivo model of vertical augmentation on rat femoral cortical bone, the interfacial strength between innate cortical bone and UV-treated microfiber scaffold after two weeks of healing was double that observed between bone and untreated scaffold. Morphological and chemical analysis confirmed seamless integration of the innate cortical and regenerated bone within microfiber networks for UV-treated scaffolds. These results indicate synergy between titanium microfiber scaffolds and UV photofunctionalization to provide a novel and effective strategy for vertical bone augmentation.
Collapse
Affiliation(s)
- Hiroaki Kitajima
- Division of Regenerative and Reconstructive Sciences and Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095-1668, USA
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Kanagawa, Japan
| | - Makoto Hirota
- Division of Regenerative and Reconstructive Sciences and Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095-1668, USA
- Department of Oral and Maxillofacial Surgery/Orthodontics, Yokohama City University Medical Center, 4-57 Urafune-cho, Minami-ku, Yokohama 236-0004, Kanagawa, Japan
- Correspondence: ; Tel./Fax: +81-45-785-8438
| | - Keiji Komatsu
- Division of Regenerative and Reconstructive Sciences and Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095-1668, USA
| | - Hitoshi Isono
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Kanagawa, Japan
| | - Takanori Matsuura
- Division of Regenerative and Reconstructive Sciences and Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095-1668, USA
| | - Kenji Mitsudo
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fuku-ura, Kanazawa-ku, Yokohama 236-0004, Kanagawa, Japan
| | - Takahiro Ogawa
- Division of Regenerative and Reconstructive Sciences and Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095-1668, USA
| |
Collapse
|
21
|
Ghuloum FI, Johnson CA, Riobo-Del Galdo NA, Amer MH. From mesenchymal niches to engineered in vitro model systems: Exploring and exploiting biomechanical regulation of vertebrate hedgehog signalling. Mater Today Bio 2022; 17:100502. [PMID: 36457847 PMCID: PMC9707069 DOI: 10.1016/j.mtbio.2022.100502] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Tissue patterning is the result of complex interactions between transcriptional programs and various mechanical cues that modulate cell behaviour and drive morphogenesis. Vertebrate Hedgehog signalling plays key roles in embryogenesis and adult tissue homeostasis, and is central to skeletal development and the osteogenic differentiation of mesenchymal stem cells. The expression of several components of the Hedgehog signalling pathway have been reported to be mechanically regulated in mesodermal tissue patterning and osteogenic differentiation in response to external stimulation. Since a number of bone developmental defects and skeletal diseases, such as osteoporosis, are directly linked to aberrant Hedgehog signalling, a better knowledge of the regulation of Hedgehog signalling in the mechanosensitive bone marrow-residing mesenchymal stromal cells will present novel avenues for modelling these diseases and uncover novel opportunities for extracellular matrix-targeted therapies. In this review, we present a brief overview of the key molecular players involved in Hedgehog signalling and the basic concepts of mechanobiology, with a focus on bone development and regeneration. We also highlight the correlation between the activation of the Hedgehog signalling pathway in response to mechanical cues and osteogenesis in bone marrow-derived mesenchymal stromal cells. Finally, we propose different tissue engineering strategies to apply the expanding knowledge of 3D material-cell interactions in the modulation of Hedgehog signalling in vitro for fundamental and translational research applications.
Collapse
Affiliation(s)
- Fatmah I. Ghuloum
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Colin A. Johnson
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Natalia A. Riobo-Del Galdo
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- Leeds Institute of Medical Research, Faculty of Medicine and Health, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK
| | - Mahetab H. Amer
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
22
|
Cyclophilin A Promotes Osteoblast Differentiation by Regulating Runx2. Int J Mol Sci 2022; 23:ijms23169244. [PMID: 36012517 PMCID: PMC9409320 DOI: 10.3390/ijms23169244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cyclophilin A (CypA) is a ubiquitously expressed and highly conserved protein with peptidyl-prolyl cis-trans isomerase activity that is involved in various biological activities by regulating protein folding and trafficking. Although CypA has been reported to positively regulate osteoblast differentiation, the mechanistic details remain largely unknown. In this study, we aimed to elucidate the mechanism of CypA-mediated regulation of osteoblast differentiation. Overexpression of CypA promoted osteoblast differentiation in bone morphogenic protein 4 (BMP4)-treated C2C12 cells, while knockdown of CypA inhibited osteoblast differentiation in BMP4-treated C2C12. CypA and Runx2 were shown to interact based on immunoprecipitation experiments and CypA increased Runx2 transcriptional activity in a dose-dependent manner. Our results indicate that this may be because CypA can increase the DNA binding affinity of Runx2 to Runx2 binding sites such as osteoblast-specific cis-acting element 2. Furthermore, to identify factors upstream of CypA in the regulation of osteoblast differentiation, various kinase inhibitors known to affect osteoblast differentiation were applied during osteogenesis. Akt inhibition resulted in the most significant suppression of osteogenesis in BMP4-induced C2C12 cells overexpressing CypA. Taken together, our results show that CypA positively regulates osteoblast differentiation by increasing the DNA binding affinity of Runx2, and Akt signaling is upstream of CypA.
Collapse
|
23
|
Multiple roles of Runt-related transcription factor-2 in tooth eruption: bone formation and resorption. Arch Oral Biol 2022; 141:105484. [PMID: 35749976 DOI: 10.1016/j.archoralbio.2022.105484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The aim was to provide a comprehensive review of the current knowledge of the multiple roles of Runt-related transcription factor-2 (RUNX2) in regulating tooth eruption, focusing on the molecular mechanisms regarding tooth eruption mediated by RUNX2. DESIGN Relevant literatures in PubMed, Medline, and Scopus database were searched, and a narrative review was performed. The multiple roles of RUNX2 in regulating tooth eruption was reviewed and discussed. RESULTS Aberrant RUNX2 expression leads to disturbed or failed tooth eruption. Tooth eruption involves both the process of bone formation and bone resorption. RUNX2 promotes osteogenesis around the radicular portion of the dental follicle that provides the biological force for tooth eruption through inducing the expression of osteogenesis-related genes in dental follicle cells/osteoblasts. On the other hand, through indirect and direct pathways, RUNX2 regulates osteoclastogenesis and the formation of the eruption pathway. CONCLUSION RUNX2 exerts a pivotal and complex influence in regulating tooth eruption. This review provides a better understanding of the function of RUNX2 in tooth eruption, which is beneficial to illuminate the precise molecular mechanism of osteogenesis and bone resorption, aiding the development of effective therapy for the failure of tooth eruption.
Collapse
|
24
|
Boyan BD, Asmussen NC, Lin Z, Schwartz Z. The Role of Matrix-Bound Extracellular Vesicles in the Regulation of Endochondral Bone Formation. Cells 2022; 11:1619. [PMID: 35626656 PMCID: PMC9139584 DOI: 10.3390/cells11101619] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 02/04/2023] Open
Abstract
Matrix vesicles are key players in the development of the growth plate during endochondral bone formation. They are involved in the turnover of the extracellular matrix and its mineralization, as well as being a vehicle for chondrocyte communication and regulation. These extracellular organelles are released by the cells and are anchored to the matrix via integrin binding to collagen. The exact function and makeup of the vesicles are dependent on the zone of the growth plate in which they are produced. Early studies defined their role as sites of initial calcium phosphate deposition based on the presence of crystals on the inner leaflet of the membrane and subsequent identification of enzymes, ion transporters, and phospholipid complexes involved in mineral formation. More recent studies have shown that they contain small RNAs, including microRNAs, that are distinct from the parent cell, raising the hypothesis that they are a distinct subset of exosomes. Matrix vesicles are produced under complex regulatory pathways, which include the action of steroid hormones. Once in the matrix, their maturation is mediated by the action of secreted hormones. How they convey information to cells, either through autocrine or paracrine actions, is now being elucidated.
Collapse
Affiliation(s)
- Barbara D. Boyan
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Niels C. Asmussen
- School of Integrated Life Sciences, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Zhao Lin
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Zvi Schwartz
- Department of Biomedical Engineering, College of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
25
|
Man K, Joukhdar H, Manz XD, Brunet MY, Jiang LH, Rnjak-Kovacina J, Yang XB. Bone tissue engineering using 3D silk scaffolds and human dental pulp stromal cells epigenetic reprogrammed with the selective histone deacetylase inhibitor MI192. Cell Tissue Res 2022; 388:565-581. [PMID: 35362831 PMCID: PMC9110470 DOI: 10.1007/s00441-022-03613-0] [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: 12/08/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022]
Abstract
Epigenetics plays a critical role in regulating mesenchymal stem cells’ (MSCs) fate for tissue repair and regeneration. There is increasing evidence that the inhibition of histone deacetylase (HDAC) isoform 3 can enhance MSC osteogenesis. This study investigated the potential of using a selective HDAC2 and 3 inhibitor, MI192, to promote human dental pulp stromal cells (hDPSCs) bone-like tissue formation in vitro and in vivo within porous Bombyx Mori silk scaffolds. Both 2 and 5 wt% silk scaffolds were fabricated and characterised. The 5 wt% scaffolds possess thicker internal lamellae, reduced scaffold swelling and degradation rates, whilst increased compressive modulus in comparison to the 2 wt% silk scaffold. MI192 pre-treatment of hDPSCs on 5 wt% silk scaffold significantly enhanced hDPSCs alkaline phosphatase activity (ALP). The expression of osteoblast-related genes (RUNX2, ALP, Col1a, OCN) was significantly upregulated in the MI192 pre-treated cells. Histological analysis confirmed that the MI192 pre-treated hDPSCs-silk scaffold constructs promoted bone extracellular matrix (ALP, Col1a, OCN) deposition and mineralisation compared to the untreated group. Following 6 weeks of subcutaneous implantation in nude mice, the MI192 pre-treated hDPSCs-silk scaffold constructs enhanced the vascularisation and extracellular matrix mineralisation compared to untreated control. In conclusion, these findings demonstrate the potential of using epigenetic reprogramming and silk scaffolds to promote hDPSCs bone formation efficacy, which provides evidence for clinical translation of this technology for bone augmentation.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.,School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Habib Joukhdar
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xue D Manz
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia.,Department of Pulmonary Medicine, Amsterdam UMC, VU University Medical Centre, Amsterdam, The Netherlands
| | - Mathieu Y Brunet
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Xuebin B Yang
- Biomaterials & Tissue Engineering Group, School of Dentistry, University of Leeds, WTBB, St. James's University Hospital, Leeds, LS97TF, UK.
| |
Collapse
|
26
|
Song JE, Lee DH, Choi JH, Lee SW, Khang G, Yoon SJ. Biomimetic sponge using duck's feet derived collagen and hydroxyapatite to promote bone regeneration. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:769-782. [PMID: 34913857 DOI: 10.1080/09205063.2021.2019366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 06/14/2023]
Abstract
Collagen, a natural biomaterial derived from animal tissues, has attracted the attention of biomedical material researchers because of its excellent cell affinity and low rejection in vivo. In this study, collagen was extracted using livestock by-product flippers, and an experiment was performed to assess its application as a scaffold for bone tissue implantation. For this purpose, we fabricated 2%, and 3% duck's feet derived collagen (DC) sponges. We then compared them to hydroxyapatite (HAp)-coated DC sponges, and measured the porosity and pore size using scanning electron microscopy (SEM) to analyze the physical properties and morphology of DC and DC/HAp sponges. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay were carried out to measure the proliferation of bone marrow stem cells (BMSCs) in DC and DC/HAp sponges. An alkaline phosphatase activity assay confirmed the osteogenic differentiation ability of BMSCs. Polymerase chain reaction (PCR) was performed to confirm the BMSC-specific genetic marker. The osteogenic potential was confirmed by the bone formation in an in vivo environment on the scaffold by histological and immunohistochemical analysis. Overall, this study shows that DC/HAp sponges have biocompatibility and good physical properties. Additionally, DC/HAp sponges show potential use as bone graft materials for tissue engineering applications.
Collapse
Affiliation(s)
- Jeong Eun Song
- Department of Bionanotechnology and Bio-Convergence Technology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Dae Hoon Lee
- Department of Bionanotechnology and Bio-Convergence Technology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Joo Hee Choi
- Department of Bionanotechnology and Bio-Convergence Technology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Seong Won Lee
- Department of Bionanotechnology and Bio-Convergence Technology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Gilson Khang
- Department of Bionanotechnology and Bio-Convergence Technology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- Department of PolymerNano Science & Technology and Polymer Materials Fusion Research Center, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Sun-Jung Yoon
- Department of PolymerNano Science & Technology and Polymer Materials Fusion Research Center, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- Department of Orthopedic Surgery, Medical School, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, South Korea
| |
Collapse
|
27
|
De la Fuente-Hernandez MA, Sarabia-Sanchez MA, Melendez-Zajgla J, Maldonado-Lagunas V. Role of lncRNAs into Mesenchymal Stromal Cell Differentiation. Am J Physiol Cell Physiol 2022; 322:C421-C460. [PMID: 35080923 DOI: 10.1152/ajpcell.00364.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Currently, findings support that 75% of the human genome is actively transcribed, but only 2% is translated into a protein, according to databases such as ENCODE (Encyclopedia of DNA Elements) [1]. The development of high-throughput sequencing technologies, computational methods for genome assembly and biological models have led to the realization of the importance of the previously unconsidered non-coding fraction of the genome. Along with this, noncoding RNAs have been shown to be epigenetic, transcriptional and post-transcriptional regulators in a large number of cellular processes [2]. Within the group of non-coding RNAs, lncRNAs represent a fascinating field of study, given the functional versatility in their mode of action on their molecular targets. In recent years, there has been an interest in learning about lncRNAs in MSC differentiation. The aim of this review is to address the signaling mechanisms where lncRNAs are involved, emphasizing their role in either stimulating or inhibiting the transition to differentiated cell. Specifically, the main types of MSC differentiation are discussed: myogenesis, osteogenesis, adipogenesis and chondrogenesis. The description of increasingly new lncRNAs reinforces their role as players in the well-studied field of MSC differentiation, allowing a step towards a better understanding of their biology and their potential application in the clinic.
Collapse
Affiliation(s)
- Marcela Angelica De la Fuente-Hernandez
- Facultad de Medicina, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sanchez
- Facultad de Medicina, Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | |
Collapse
|
28
|
Xie F, Cui QK, Wang ZY, Liu B, Qiao W, Li N, Cheng J, Hou YM, Dong XY, Wang Y, Zhang MX. ILF3 is responsible for hyperlipidemia-induced arteriosclerotic calcification by mediating BMP2 and STAT1 transcription. J Mol Cell Cardiol 2021; 161:39-52. [PMID: 34343541 DOI: 10.1016/j.yjmcc.2021.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/01/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022]
Abstract
Calcification is common in atherosclerotic plaque and can induce vulnerability, which further leads to myocardial infarction, plaque rupture and stroke. The mechanisms of atherosclerotic calcification are poorly characterized. Interleukin enhancer binding factor 3 (ILF3) has been identified as a novel factor affecting dyslipidemia and stroke subtypes. However, the precise role of ILF3 in atherosclerotic calcification remains unclear. In this study, we used smooth muscle-conditional ILF3 knockout (ILF3SM-KO) and transgenic mice (ILF3SM-Tg) and macrophage-conditional ILF3 knockout (ILF3M-KO) and transgenic (ILF3M-Tg) mice respectively. Here we showed that ILF3 expression is increased in calcified human aortic vascular smooth muscle cells (HAVSMCs) and calcified atherosclerotic plaque in humans and mice. We then found that hyperlipidemia increases ILF3 expression and exacerbates calcification of VSMCs and macrophages by regulating bone morphogenetic protein 2 (BMP2) and signal transducer and activator of transcription 1 (STAT1) transcription. We further explored the molecular mechanisms of ILF3 in atherosclerotic calcification and revealed that ILF3 acts on the promoter regions of BMP2 and STAT1 and mediates BMP2 upregulation and STAT1 downregulation, which promotes atherosclerotic calcification. Our results demonstrate the effect of ILF3 in atherosclerotic calcification. Inhibition of ILF3 may be a useful therapy for preventing and even reversing atherosclerotic calcification.
Collapse
Affiliation(s)
- Fei Xie
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qing-Ke Cui
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Zhao-Yang Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bin Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wen Qiao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Na Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ya-Min Hou
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xin-Ying Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ying Wang
- Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Ming-Xiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
29
|
Man K, Mekhileri NV, Lim KS, Jiang LH, Woodfield TBF, Yang XB. MI192 induced epigenetic reprogramming enhances the therapeutic efficacy of human bone marrows stromal cells for bone regeneration. Bone 2021; 153:116138. [PMID: 34339909 DOI: 10.1016/j.bone.2021.116138] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
Human bone marrow stromal cells (hBMSCs) have been extensively utilised for bone tissue engineering applications. However, they are associated with limitations that hinder their clinical utility for bone regeneration. Cell fate can be modulated via altering their epigenetic functionality. Inhibiting histone deacetylase (HDAC) enzymes have been reported to promote osteogenic differentiation, with HDAC3 activity shown to be causatively associated with osteogenesis. Therefore, this study aimed to investigate the potential of using an HDAC2 & 3 selective inhibitor - MI192 to induce epigenetic reprogramming of hBMSCs and enhance its therapeutic efficacy for bone formation. Treatment with MI192 caused a time-dose dependant reduction in hBMSCs viability. MI192 was also found to substantially alter hBMSCs epigenetic function through reduced HDAC activity and increased histone acetylation. hBMSCs were pre-treated with MI192 (50 μM) for 48 h prior to osteogenic induction. MI192 pre-treatment significantly upregulated osteoblast-related gene/protein expression (Runx2, ALP, Col1a and OCN) and enhanced alkaline phosphatase specific activity (ALPSA) (1.43-fold) (P ≤ 0.001). Moreover, MI192 substantially increased hBMSCs extracellular matrix calcium deposition (1.4-fold) (P ≤ 0.001) and mineralisation when compared to the untreated control. In 3D microtissue culture, MI192 significantly promoted hBMSCs osteoblast-related gene expression and ALPSA (> 2.41-fold) (P ≤ 0.001). Importantly, MI192 substantially enhanced extracellular matrix deposition (ALP, Col1a, OCN) and mineralisation (1.67-fold) (P ≤ 0.001) within the bioassembled-microtissue (BMT) construct. Following 8-week intraperitoneal implantation within nude mice, MI192 treated hBMSCs exhibited enhanced extracellular matrix deposition and mineralisation (2.39-fold) (P ≤ 0.001) within the BMT when compared to the untreated BMT construct. Taken together, these results demonstrate that MI192 effectively altered hBMSCs epigenetic functionality and is capable of promoting hBMSCs osteogenic differentiation in vitro and in vivo, indicating the potential of using epigenetic reprogramming to enhance the therapeutic efficacy of hBMSCs for bone augmentation strategies.
Collapse
Affiliation(s)
- Kenny Man
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK; School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Naveen V Mekhileri
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Lin-Hua Jiang
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Tim B F Woodfield
- CReaTE Group, Department of Orthopaedic Surgery, University of Otago, Christchurch, New Zealand
| | - Xuebin B Yang
- Biomaterial and Tissue Engineering Group, School of Dentistry, University of Leeds, Leeds, UK.
| |
Collapse
|
30
|
Mechanical and Biological Properties of Magnesium- and Silicon-Substituted Hydroxyapatite Scaffolds. MATERIALS 2021; 14:ma14226942. [PMID: 34832344 PMCID: PMC8619624 DOI: 10.3390/ma14226942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022]
Abstract
Magnesium (Mg)- and silicon (Si)-substituted hydroxyapatite (HA) scaffolds were synthesized using the sponge replica method. The influence of Mg2+ and SiO44− ion substitution on the microstructural, mechanical and biological properties of HA scaffolds was evaluated. All synthesized scaffolds exhibited porosity >92%, with interconnected pores and pore sizes ranging between 200 and 800 μm. X-ray diffraction analysis showed that β-TCP was formed in the case of Mg substitution. X-ray fluorescence mapping showed a homogeneous distribution of Mg and Si ions in the respective scaffolds. Compared to the pure HA scaffold, a reduced grain size was observed in the Mg- and Si-substituted scaffolds, which greatly influenced the mechanical properties of the scaffolds. Mechanical tests revealed better performance in HA-Mg (0.44 ± 0.05 MPa), HA-Si (0.64 ± 0.02 MPa) and HA-MgSi (0.53 ± 0.01 MPa) samples compared to pure HA (0.2 ± 0.01 MPa). During biodegradability tests in Tris-HCl, slight weight loss and a substantial reduction in mechanical performances of the scaffolds were observed. Cell proliferation determined by the MTT assay using hBMSC showed that all scaffolds were biocompatible, and the HA-MgSi scaffold seemed the most effective for cell adhesion and proliferation. Furthermore, ALP activity and osteogenic marker expression analysis revealed the ability of HA-Si and HA-MgSi scaffolds to promote osteoblast differentiation.
Collapse
|
31
|
NaF reduces KLK4 expression by decreasing Foxo1/Runx2 expression in LS8 cells. Arch Oral Biol 2021; 133:105311. [PMID: 34781073 DOI: 10.1016/j.archoralbio.2021.105311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/19/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the effect of high fluoride on runt-related transcription factor 2 (Runx2) expression and to explore the possible relationship among Runx2, forkhead box o1 (Foxo1) and kallikrein 4 (KLK4) in high fluoride-treated ameloblasts. DESIGN Ameloblast-like cells (LS8 cells) were exposed to various concentrations of sodium fluoride (NaF) for up to 48 h. Runx2 expression was downregulated by gene silencing, and Foxo1 expression was up- and downregulated by gene overexpression and silencing, respectively. The mRNA and protein levels of Runx2, Foxo1, KLK4 and matrix metalloproteinase 20 (MMP20) were detected by qRT-PCR and western blotting. RESULTS Runx2 expression was decreased in a dose- and time-dependent manner in NaF-treated LS8 cells. The knockdown of Runx2 markedly decreased KLK4 expression in LS8 cells under NaF conditions. However, the variation trend of MMP20 was unclear. In addition, forced Foxo1 expression led to significant upregulation of Runx2 in LS8 cells under NaF conditions. In contrast, the knockdown of Foxo1 markedly decreased the Runx2 protein levels under NaF conditions. Moreover, Foxo1 downregulation markedly decreased runx2 mRNA levels, and this inhibition in LS8 cells was intensified when combined with NaF treatment. CONCLUSION The results indicated that NaF reduces Runx2 expression in LS8 cells and that decreased Foxo1/Runx2 expression induced by high fluoride is a cause of low KLK4 expression.
Collapse
|
32
|
Masbuchin AN, Rohman MS, Liu PY. Role of Glycosylation in Vascular Calcification. Int J Mol Sci 2021; 22:9829. [PMID: 34575990 PMCID: PMC8469761 DOI: 10.3390/ijms22189829] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Glycosylation is an important step in post-translational protein modification. Altered glycosylation results in an abnormality that causes diseases such as malignancy and cardiovascular diseases. Recent emerging evidence highlights the importance of glycosylation in vascular calcification. Two major types of glycosylation, N-glycosylation and O-glycosylation, are involved in vascular calcification. Other glycosylation mechanisms, which polymerize the glycosaminoglycan (GAG) chain onto protein, resulting in proteoglycan (PG), also have an impact on vascular calcification. This paper discusses the role of glycosylation in vascular calcification.
Collapse
Affiliation(s)
- Ainun Nizar Masbuchin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan;
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang 65111, Indonesia;
| | - Mohammad Saifur Rohman
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Brawijaya, Malang 65111, Indonesia;
| | - Ping-Yen Liu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan;
- Division of Cardiology, Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| |
Collapse
|
33
|
Man K, Brunet MY, Fernandez‐Rhodes M, Williams S, Heaney LM, Gethings LA, Federici A, Davies OG, Hoey D, Cox SC. Epigenetic reprogramming enhances the therapeutic efficacy of osteoblast-derived extracellular vesicles to promote human bone marrow stem cell osteogenic differentiation. J Extracell Vesicles 2021; 10:e12118. [PMID: 34262674 PMCID: PMC8263905 DOI: 10.1002/jev2.12118] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/18/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are emerging in tissue engineering as promising acellular tools, circumventing many of the limitations associated with cell-based therapies. Epigenetic regulation through histone deacetylase (HDAC) inhibition has been shown to increase differentiation capacity. Therefore, this study aimed to investigate the potential of augmenting osteoblast epigenetic functionality using the HDAC inhibitor Trichostatin A (TSA) to enhance the therapeutic efficacy of osteoblast-derived EVs for bone regeneration. TSA was found to substantially alter osteoblast epigenetic function through reduced HDAC activity and increased histone acetylation. Treatment with TSA also significantly enhanced osteoblast alkaline phosphatase activity (1.35-fold), collagen production (2.8-fold) and calcium deposition (1.55-fold) during osteogenic culture (P ≤ 0.001). EVs derived from TSA-treated osteoblasts (TSA-EVs) exhibited reduced particle size (1-05-fold) (P > 0.05), concentration (1.4-fold) (P > 0.05) and protein content (1.16-fold) (P ≤ 0.001) when compared to untreated EVs. TSA-EVs significantly enhanced the proliferation (1.13-fold) and migration (1.3-fold) of human bone marrow stem cells (hBMSCs) when compared to untreated EVs (P ≤ 0.05). Moreover, TSA-EVs upregulated hBMSCs osteoblast-related gene and protein expression (ALP, Col1a, BSP1 and OCN) when compared to cells cultured with untreated EVs. Importantly, TSA-EVs elicited a time-dose dependent increase in hBMSCs extracellular matrix mineralisation. MicroRNA profiling revealed a set of differentially expressed microRNAs from TSA-EVs, which were osteogenic-related. Target prediction demonstrated these microRNAs were involved in regulating pathways such as 'endocytosis' and 'Wnt signalling pathway'. Moreover, proteomics analysis identified the enrichment of proteins involved in transcriptional regulation within TSA-EVs. Taken together, our findings suggest that altering osteoblasts' epigenome accelerates their mineralisation and promotes the osteoinductive potency of secreted EVs partly due to the delivery of pro-osteogenic microRNAs and transcriptional regulating proteins. As such, for the first time we demonstrate the potential to harness epigenetic regulation as a novel engineering approach to enhance EVs therapeutic efficacy for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| | | | | | - Soraya Williams
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Liam M. Heaney
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - Lee A. Gethings
- Waters CorporationStamford AvenueWilmslowUK
- Division of Infection, Immunity and Respiratory MedicineFaculty of Biology, Medicine and HealthManchester Institute of BiotechnologyUniversity of ManchesterManchesterUK
| | - Angelica Federici
- Trinity Biomedical Sciences InstituteTrinity CollegeTrinity Centre for Biomedical EngineeringDublinIreland
- Department of Mechanical, Manufacturing, and Biomedical EngineeringSchool of EngineeringTrinity College DublinIreland
- Trinity College Dublin & RCSIAdvanced Materials and Bioengineering Research CentreDublinIreland
| | - Owen G. Davies
- School of Sport, Exercise and Health SciencesLoughborough UniversityLoughboroughUK
| | - David Hoey
- Trinity Biomedical Sciences InstituteTrinity CollegeTrinity Centre for Biomedical EngineeringDublinIreland
- Department of Mechanical, Manufacturing, and Biomedical EngineeringSchool of EngineeringTrinity College DublinIreland
- Trinity College Dublin & RCSIAdvanced Materials and Bioengineering Research CentreDublinIreland
| | - Sophie C. Cox
- School of Chemical EngineeringUniversity of BirminghamBirminghamUK
| |
Collapse
|
34
|
PP2A in LepR+ mesenchymal stem cells contributes to embryonic and postnatal endochondral ossification through Runx2 dephosphorylation. Commun Biol 2021; 4:658. [PMID: 34079065 PMCID: PMC8172534 DOI: 10.1038/s42003-021-02175-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
It has not been well studied which cells and related mechanisms contribute to endochondral ossification. Here, we fate mapped the leptin receptor-expressing (LepR+) mesenchymal stem cells (MSCs) in different embryonic and adult extremities using Lepr-cre; tdTomato mice and investigated the underling mechanism using Lepr-cre; Ppp2r1afl/fl mice. Tomato+ cells appear in the primary and secondary ossification centers and express the hypertrophic markers. Ppp2r1a deletion in LepR+ MSCs reduces the expression of Runx2, Osterix, alkaline phosphatase, collagen X, and MMP13, but increases that of the mature adipocyte marker perilipin, thereby reducing trabecular bone density and enhancing fat content. Mechanistically, PP2A dephosphorylates Runx2 and BRD4, thereby playing a major role in positively and negatively regulating osteogenesis and adipogenesis, respectively. Our data identify LepR+ MSC as the cell origin of endochondral ossification during embryonic and postnatal bone growth and suggest that PP2A is a therapeutic target in the treatment of dysregulated bone formation. Yen et al use tissue-specific PP2A knockout mice to show that PP2A in LepR positive mesenchymal stem cells positively regulates endochondral ossification. They find that PP2A dephosphorylates Runx2 and BRD4, thereby playing a major role in positively and negatively regulating osteogenesis and adipogenesis, respectively.
Collapse
|
35
|
The Selective Histone Deacetylase Inhibitor MI192 Enhances the Osteogenic Differentiation Efficacy of Human Dental Pulp Stromal Cells. Int J Mol Sci 2021; 22:ijms22105224. [PMID: 34069280 PMCID: PMC8156347 DOI: 10.3390/ijms22105224] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
The use of human dental pulp stromal cells (hDPSCs) has gained increasing attention as an alternative stem cell source for bone tissue engineering. The modification of the cells' epigenetics has been found to play an important role in regulating differentiation, with the inhibition of histone deacetylases 3 (HDAC3) being linked to increased osteogenic differentiation. This study aimed to induce epigenetic reprogramming using the HDAC2 and 3 selective inhibitor, MI192 to promote hDPSCs osteogenic capacity for bone regeneration. MI192 treatment caused a time-dose-dependent change in hDPSC morphology and reduction in viability. Additionally, MI192 successfully augmented hDPSC epigenetic functionality, which resulted in increased histone acetylation and cell cycle arrest at the G2/M phase. MI192 pre-treatment exhibited a dose-dependent effect on hDPSCs alkaline phosphatase activity. Quantitative PCR and In-Cell Western further demonstrated that MI192 pre-treatment significantly upregulated hDPSCs osteoblast-related gene and protein expression (alkaline phosphatase, bone morphogenic protein 2, type I collagen and osteocalcin) during osteogenic differentiation. Importantly, MI192 pre-treatment significantly increased hDPSCs extracellular matrix collagen production and mineralisation. As such, for the first time, our findings show that epigenetic reprogramming with the HDAC2 and 3 selective inhibitor MI192 accelerates the osteogenic differentiation of hDPSCs, demonstrating the considerable utility of this MSCs engineering approach for bone augmentation strategies.
Collapse
|
36
|
Lanzillotti C, De Mattei M, Mazziotta C, Taraballi F, Rotondo JC, Tognon M, Martini F. Long Non-coding RNAs and MicroRNAs Interplay in Osteogenic Differentiation of Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:646032. [PMID: 33898434 PMCID: PMC8063120 DOI: 10.3389/fcell.2021.646032] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/11/2021] [Indexed: 12/23/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have gained great attention as epigenetic regulators of gene expression in many tissues. Increasing evidence indicates that lncRNAs, together with microRNAs (miRNAs), play a pivotal role in osteogenesis. While miRNA action mechanism relies mainly on miRNA-mRNA interaction, resulting in suppressed expression, lncRNAs affect mRNA functionality through different activities, including interaction with miRNAs. Recent advances in RNA sequencing technology have improved knowledge into the molecular pathways regulated by the interaction of lncRNAs and miRNAs. This review reports on the recent knowledge of lncRNAs and miRNAs roles as key regulators of osteogenic differentiation. Specifically, we described herein the recent discoveries on lncRNA-miRNA crosstalk during the osteogenic differentiation of mesenchymal stem cells (MSCs) derived from bone marrow (BM), as well as from different other anatomical regions. The deep understanding of the connection between miRNAs and lncRNAs during the osteogenic differentiation will strongly improve knowledge into the molecular mechanisms of bone growth and development, ultimately leading to discover innovative diagnostic and therapeutic tools for osteogenic disorders and bone diseases.
Collapse
Affiliation(s)
- Carmen Lanzillotti
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Monica De Mattei
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Chiara Mazziotta
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX, United States.,Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - John Charles Rotondo
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| |
Collapse
|
37
|
Liu X, Li X, Hua B, Yang X, Zheng J, Liu S. WNT16 is upregulated early in mouse TMJ osteoarthritis and protects fibrochondrocytes against IL-1β induced inflammatory response by regulation of RUNX2/MMP13 cascade. Bone 2021; 143:115793. [PMID: 33301961 DOI: 10.1016/j.bone.2020.115793] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/30/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023]
Abstract
WNT16 has been shown to play important roles in joint formation, bone homeostasis and knee joint osteoarthritis. However, whether WNT16 has any effect during temporomandibular joint osteoarthritis (TMJOA) is still unknown. Here, we first established a surgically induced TMJOA model by performing partial discectomy in discs of TMJ in mice. Further, we investigated the role of WNT16 during the initiation and progression of TMJOA. Our results showed that WNT16 expression is upregulated early at 4 weeks after initiation of osteoarthritis by partial discectomy in mouse TMJ cartilage, but decreased after 12 weeks post-surgery. Further cellular and molecular analyses revealed that WNT16 signals via both the canonical WNT/β-catenin and non-canonical WNT/JNK-cJUN pathways, upregulates the expression of Lubricin and SOX9, and protects against IL-1β induced inflammatory response by regulation of RUNX2/MMP13 cascade in fibrochondrocytes. In conclusion, WNT16 may play an important role in the early stage of TMJOA by regulating cartilage anabolic and catabolic factors, and may serve as novel therapeutic targets for TMJOA.
Collapse
Affiliation(s)
- Xianwen Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xinping Li
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Bingqiang Hua
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoqin Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Junfa Zheng
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| | - Shuguang Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Yamada S, Yassin MA, Schwarz T, Hansmann J, Mustafa K. Induction of osteogenic differentiation of bone marrow stromal cells on 3D polyester-based scaffolds solely by subphysiological fluidic stimulation in a laminar flow bioreactor. J Tissue Eng 2021; 12:20417314211019375. [PMID: 34262684 PMCID: PMC8243246 DOI: 10.1177/20417314211019375] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023] Open
Abstract
The fatal determination of bone marrow mesenchymal stem/stromal cells (BMSC) is closely associated with mechano-environmental factors in addition to biochemical clues. The aim of this study was to induce osteogenesis in the absence of chemical stimuli using a custom-designed laminar flow bioreactor. BMSC were seeded onto synthetic microporous scaffolds and subjected to the subphysiological level of fluid flow for up to 21 days. During the perfusion, cell proliferation was significantly inhibited. There were also morphological changes, with F-actin polymerisation and upregulation of ROCK1. Notably, in BMSC subjected to flow, mRNA expression of osteogenic markers was significantly upregulated and RUNX2 was localised in the nuclei. Further, under perfusion, there was greater deposition of collagen type 1 and calcium onto the scaffolds. The results confirm that an appropriate level of fluid stimuli preconditions BMSC towards the osteoblastic lineage on 3D scaffolds in the absence of chemical stimulation, which highlights the utility of flow bioreactors in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Mohammed Ahmed Yassin
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
- Chair of Tissue Engineering and
Regenerative Medicine, University Hospital Würzburg, Germany
- Department Electrical Engineering,
University of Applied Sciences Würzburg-Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| |
Collapse
|
39
|
Brito VGB, Patrocinio MS, de Sousa MCL, Barreto AEA, Frasnelli SCT, Lara VS, Santos CF, Oliveira SHP. Telmisartan Prevents Alveolar Bone Loss by Decreasing the Expression of Osteoclasts Markers in Hypertensive Rats With Periodontal Disease. Front Pharmacol 2020; 11:579926. [PMID: 33364953 PMCID: PMC7751694 DOI: 10.3389/fphar.2020.579926] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022] Open
Abstract
Periodontal disease (PD) is a prevalent inflammatory disease with the most severe consequence being the loss of the alveolar bone and teeth. We therefore aimed to evaluate the effects of telmisartan (TELM), an angiotensin II type 1 receptor (Agtr1) antagonist, on the PD-induced alveolar bone loss, in Wistar (W) and Spontaneous Hypertensive Rats (SHRs). PD was induced by ligating the lower first molars with silk, and 10 mg/kg TELM was concomitantly administered for 15 days. The hemimandibles were subjected to microtomography, ELISA was used for detecting tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), CXCL3, and CCL2, while qRT-PCR was used for analyzing expression of components of renin-angiotensin system (RAS) (Agt, Ace, Agt1r, Agt2r, Ace2, and Masr), and bone markers (Runx2, Osx, Catnb, Alp, Col1a1, Opn, Ocn, Bsp, Bmp2, Trap, Rank, Rankl, CtsK, Mmp-2, Mmp-9, and osteoclast-associated receptor (Oscar)). The SHR + PD group showed greater alveolar bone loss than the W + PD group, what was significantly inhibited by treatment with TELM, especially in the SHR group. Additionally, TELM reduced the production of TNF-α, IL-1β, and CXCL3 in the SHR group. The expression of Agt increased in the groups with PD, while Agtr2 reduced, and TELM reduced the expression of Agtr1 and increased the expression of Agtr2, in W and SHRs. PD did not induce major changes in the expression of bone formation markers, except for the expression of Alp, which decreased in the PD groups. The bone resorption markers expression, Mmp9, Ctsk, and Vtn, was higher in the SHR + PD group, compared to the respective control and W + PD group. However, TELM attenuated these changes and increased the expression of Runx2 and Alp. Our study suggested that TELM has a protective effect on the progression of PD, especially in hypertensive animals, as evaluated by the resorption of the lower alveolar bone. This can be partly explained by the modulation in the expression of Angiotensin II receptors (AT1R and AT2R), reduced production of inflammatory mediators, the reduced expression of resorption markers, and the increased expression of the bone formation markers.
Collapse
Affiliation(s)
- Victor Gustavo Balera Brito
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Mariana Sousa Patrocinio
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Maria Carolina Linjardi de Sousa
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Ayná Emanuelli Alves Barreto
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Sabrina Cruz Tfaile Frasnelli
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| | - Vanessa Soares Lara
- Department of Stomatology, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Carlos Ferreira Santos
- Department of Biological Science, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Sandra Helena Penha Oliveira
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil.,Multicenter Postgraduate Program in Physiological Sciences, Brazilian Society of Physiology, School of Dentistry, São Paulo State University (UNESP), Araçatuba, Brazil
| |
Collapse
|
40
|
Concentrated Growth Factors (CGF) Induce Osteogenic Differentiation in Human Bone Marrow Stem Cells. BIOLOGY 2020; 9:biology9110370. [PMID: 33143015 PMCID: PMC7693660 DOI: 10.3390/biology9110370] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022]
Abstract
Bone regeneration is a complex process regulated by several factors that control overlapping biological processes, coordinating interactions among distinct cell populations. There is a great interest in identifying new strategies for inducing osteogenesis in a safe and efficient manner. Concentrated Growth Factor (CGF) is an autologous blood derived product obtained by centrifugation of venous blood following the procedure set on the Silfradent device. In this study the effects of CGF on osteogenic differentiation of human Bone Marrow Stem Cells (hBMSC) in vitro have been investigated; hBMSC were cultured with CGF or osteogenic medium, for 21 days. The osteogenic differentiation was evaluated measuring alkaline phosphatase (ALP) enzyme activity, matrix mineralization by alizarin red staining and through mRNA and protein quantification of osteogenic differentiation markers by Real-time PCR and Western blotting, respectively. The treatment with CGF stimulated ALP activity and promoted matrix mineralization compared to control and seems to be more effective than osteogenic medium. Also, hBMSC lost mesenchymal markers and showed other osteogenic features. Our study showed for the first time that CGF alone is able to induce osteogenic differentiation in hBMSC. The application of CGF on hBMSC osteoinduction might offer new clinical and biotechnological strategies in the tissue regeneration field.
Collapse
|
41
|
Dai G, Xiao H, Zhao C, Chen H, Liao J, Huang W. LncRNA H19 Regulates BMP2-Induced Hypertrophic Differentiation of Mesenchymal Stem Cells by Promoting Runx2 Phosphorylation. Front Cell Dev Biol 2020; 8:580. [PMID: 32903671 PMCID: PMC7438821 DOI: 10.3389/fcell.2020.00580] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Objectives Bone morphogenetic protein 2 (BMP2) triggers hypertrophic differentiation after chondrogenic differentiation of mesenchymal stem cells (MSCs), which blocked the further application of BMP2-mediated cartilage tissue engineering. Here, we investigated the underlying mechanisms of BMP2-mediated hypertrophic differentiation of MSCs. Materials and Methods In vitro and in vivo chondrogenic differentiation models of MSCs were constructed. The expression of H19 in mouse limb was detected by fluorescence in situ hybridization (FISH) analysis. Transgenes BMP2, H19 silencing, and overexpression were expressed by adenoviral vectors. Gene expression was determined by reverse transcription and quantitative real-time PCR (RT-qPCR), Western blot, and immunohistochemistry. Correlations between H19 expressions and other parameters were calculated with Spearman’s correlation coefficients. The combination of H19 and Runx2 was identified by RNA immunoprecipitation (RIP) analysis. Results We identified that H19 expression level was highest in proliferative zone and decreased gradually from prehypertrophic zone to hypertrophic zone in mouse limbs. With the stimulation of BMP2, the highest expression level of H19 was followed after the peak expression level of Sox9; meanwhile, H19 expression levels were positively correlated with chondrogenic differentiation markers, especially in the late stage of BMP2 stimulation, and negatively correlated with hypertrophic differentiation markers. Our further experiments found that silencing H19 promoted BMP2-triggered hypertrophic differentiation through in vitro and in vivo tests, which indicated the essential role of H19 for maintaining the phenotype of BMP2-induced chondrocytes. In mechanism, we characterized that H19 regulated BMP2-mediated hypertrophic differentiation of MSCs by promoting the phosphorylation of Runx2. Conclusion These findings suggested that H19 regulates BMP2-induced hypertrophic differentiation of MSCs by promoting the phosphorylation of Runx2.
Collapse
Affiliation(s)
- Guangming Dai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haozhuo Xiao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chen Zhao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junyi Liao
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
42
|
Byon CH, Kim SW. Regulatory Effects of O-GlcNAcylation in Vascular Smooth Muscle Cells on Diabetic Vasculopathy. J Lipid Atheroscler 2020; 9:243-254. [PMID: 32821734 PMCID: PMC7379086 DOI: 10.12997/jla.2020.9.2.243] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/26/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
Vascular complications from uncontrolled hyperglycemia are the leading cause of death in patients with diabetes mellitus. Previous reports have shown a strong correlation between hyperglycemia and vascular calcification, which increases mortality and morbidity in individuals with diabetes. However, the precise underlying molecular mechanisms of hyperglycemia-induced vascular calcification remain largely unknown. Transdifferentiation of vascular smooth muscle cells (VSMC) into osteoblast-like cells is a known culprit underlying the development of vascular calcification in the diabetic vasculature. Pathological conditions such as high glucose levels and oxidative stress are linked to enhanced osteogenic differentiation of VSMC both in vivo and in vitro. It has been demonstrated that increased expression of runt-related transcription factor 2 (Runx2), a bone-related transcription factor, in VSMC is necessary and sufficient for the induction of VSMC calcification. Addition of a single O-linked β-N-acetylglucosamine (O-GlcNAc) moiety to the serine/threonine residues of target proteins (O-GlcNAcylation) has been observed in the arteries of diabetic patients, as well as in animal models in association with the enhanced expression of Runx2 and aggravated vascular calcification. O-GlcNAcylation is a dynamic and tightly regulated process, that is mediated by 2 enzymes, O-GlcNAc transferase and O-GlcNAcase. Glucose is metabolized into UDP-β-D-N-acetylglucosamine, an active sugar donor of O-GlcNAcylation via the hexosamine biosynthetic pathway. Overall increases in the O-GlcNAcylation of cellular proteins have been closely associated with cardiovascular complications of diabetes. In this review, the authors provide molecular insights into cardiovascular complications, including diabetic vasculopathy, that feature increased O-GlcNAcylation in people with diabetes.
Collapse
Affiliation(s)
- Chang Hyun Byon
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
43
|
Zhao X, Xie L, Wang Z, Wang J, Xu H, Han X, Bai D, Deng P. ZBP1 (DAI/DLM-1) promotes osteogenic differentiation while inhibiting adipogenic differentiation in mesenchymal stem cells through a positive feedback loop of Wnt/β-catenin signaling. Bone Res 2020; 8:12. [PMID: 32195010 PMCID: PMC7058036 DOI: 10.1038/s41413-020-0085-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 06/19/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023] Open
Abstract
The lineage specification of mesenchymal stem/stromal cells (MSCs) is tightly regulated by a wide range of factors. Recently, the versatile functions of ZBP1 (also known as DAI or DLM-1) have been reported in the blood circulation and immune systems. However, the biological function of ZBP1 during the lineage specification of MSCs is still unknown. In the present study, we found that ZBP1 was upregulated during osteogenesis but downregulated during adipogenesis in mouse bone marrow-derived MSCs (mBMSCs). ZBP1 was highly expressed in osteoblasts but expressed at a relatively low level in marrow adipocytes. Knockdown of ZBP1 inhibited alkaline phosphataseactivity, extracellular matrix mineralization, and osteogenesis-related gene expression in vitro and reduced ectopic bone formation in vivo. Knockdown of ZBP1 also promoted adipogenesis in MSCs in vitro. Conversely, the overexpression of ZBP1 increased the osteogenesis but suppressed the adipogenesis of MSCs. When the expression of ZBP1 was rescued, the osteogenic capacity of ZBP1-depleted mBMSCs was restored at both the molecular and phenotypic levels. Furthermore, we demonstrated that ZBP1, a newly identified target of Wnt/β-catenin signaling, was required for β-catenin translocation into nuclei. Collectively, our results indicate that ZBP1 is a novel regulator of bone and fat transdifferentiation via Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Xuefeng Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Zhiyong Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Ding Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| | - Peng Deng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041 PR China
| |
Collapse
|
44
|
Malavika D, Shreya S, Raj Priya V, Rohini M, He Z, Partridge NC, Selvamurugan N. miR‐873‐3p targets HDAC4 to stimulate matrix metalloproteinase‐13 expression upon parathyroid hormone exposure in rat osteoblasts. J Cell Physiol 2020; 235:7996-8009. [DOI: 10.1002/jcp.29454] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/03/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Desai Malavika
- Department of Biotechnology, School of Bioengineering SRM Institute of Science and Technology Kattankulathur Tamil Nadu India
| | - Srinivasan Shreya
- Department of Biotechnology, School of Bioengineering SRM Institute of Science and Technology Kattankulathur Tamil Nadu India
| | - Vembar Raj Priya
- Department of Biotechnology, School of Bioengineering SRM Institute of Science and Technology Kattankulathur Tamil Nadu India
| | - Muthukumar Rohini
- Department of Biotechnology, School of Bioengineering SRM Institute of Science and Technology Kattankulathur Tamil Nadu India
| | - Zhiming He
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry New York University New York New York
| | - Nicola C. Partridge
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry New York University New York New York
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering SRM Institute of Science and Technology Kattankulathur Tamil Nadu India
| |
Collapse
|
45
|
Galea GL, Paradise CR, Meakin LB, Camilleri ET, Taipaleenmaki H, Stein GS, Lanyon LE, Price JS, van Wijnen AJ, Dudakovic A. Mechanical strain-mediated reduction in RANKL expression is associated with RUNX2 and BRD2. Gene 2020; 763S:100027. [PMID: 32550554 PMCID: PMC7285908 DOI: 10.1016/j.gene.2020.100027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 01/08/2023]
Abstract
Mechanical loading-related strains trigger bone formation by osteoblasts while suppressing resorption by osteoclasts, uncoupling the processes of formation and resorption. Osteocytes may orchestrate this process in part by secreting sclerostin (SOST), which inhibits osteoblasts, and expressing receptor activator of nuclear factor-κB ligand (RANKL/TNFSF11) which recruits osteoclasts. Both SOST and RANKL are targets of the master osteoblastic transcription factor RUNX2. Subjecting human osteoblastic Saos-2 cells to strain by four point bending down-regulates their expression of SOST and RANKL without altering RUNX2 expression. RUNX2 knockdown increases basal SOST expression, but does not alter SOST down-regulation following strain. Conversely, RUNX2 knockdown does not alter basal RANKL expression, but prevents its down-regulation by strain. Chromatin immunoprecipitation revealed RUNX2 occupies a region of the RANKL promoter containing a consensus RUNX2 binding site and its occupancy of this site decreases following strain. The expression of epigenetic acetyl and methyl writers and readers was quantified by RT-qPCR to investigate potential epigenetic bases for this change. Strain and RUNX2 knockdown both down-regulate expression of the bromodomain acetyl reader BRD2. BRD2 and RUNX2 co-immunoprecipitate, suggesting interaction within regulatory complexes, and BRD2 was confirmed to interact with the RUNX2 promoter. BRD2 also occupies the RANKL promoter and its occupancy was reduced following exposure to strain. Thus, RUNX2 may contribute to bone remodeling by suppressing basal SOST expression, while facilitating the acute strain-induced down-regulation of RANKL through a mechanosensitive epigenetic loop involving BRD2.
Collapse
Key Words
- ALP, Alkaline phosphatase
- ActD, Actinomycin D
- AzadC, 5-Aza-2′-deoxycytidine
- BRD2
- BRD2, Bromodomain-containing protein 2
- CO2, Carbon Dioxide
- ChIP, Chromatin immunoprecipitation
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM, Dulbecco's Modified Eagle Medium
- DNA, Deoxyribonucleic Acid
- Epigenetics
- FACS, Fluorescence-activated cell sorting
- FCS, Fetal calf serum
- GAPDH, Glyceraldehyde 3-Phosphate Dehydrogenase
- HDAC, Histone deacetylase
- HPRT, Hypoxanthine Phosphoribosyltransferase 1
- IU, International unit
- IgG, Immunoglobulin G
- Ki-67, Antigen KI-67
- Mechanical strain
- OPG, Osteoprotegerin/tumour necrosis factor receptor superfamily member 11B
- PBS, Phosphate-Buffered Saline
- PCR, polymerase chain reaction
- PGE2, Prostaglandin E2
- RANKL/TNFSF11, receptor activator of nuclear factor-κB ligand
- RNA, Ribonucleic Acid
- RT-qPCR, Quantitative reverse transcription polymerase chain reaction
- RUNX2
- RUNX2, Runt-related transcription factor 2
- Receptor activator of nuclear factor-κB ligand
- SOST, Sclerostin
- Sclerostin
- eGFP, enhanced green fluorescent protein
- sh, Short hairpin
- β2MG, Beta-2-Microglobulin
Collapse
Affiliation(s)
- Gabriel L Galea
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK.,Comparative Bioveterinary Sciences, Royal Veterinary College, London, UK
| | - Christopher R Paradise
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA.,Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lee B Meakin
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | | | - Hanna Taipaleenmaki
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gary S Stein
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, VT, USA
| | - Lance E Lanyon
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Joanna S Price
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
46
|
Chen D, Kim DJ, Shen J, Zou Z, O'Keefe RJ. Runx2 plays a central role in Osteoarthritis development. J Orthop Translat 2019; 23:132-139. [PMID: 32913706 PMCID: PMC7452174 DOI: 10.1016/j.jot.2019.11.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, is the leading cause of impaired mobility in the elderly, and accounts for more than a third of chronic moderate to severe pain. As a degenerative joint disorder, OA affects the whole joint and results in synovial hyperplasia, degradation of articular cartilage, subchondral sclerosis, osteophyte formation, and chronic pain. Currently, there is no effective drug to decelerate OA progression and molecular targets for drug development have been insufficiently investigated. Anti-OA drug development can benefit from more and precise knowledge of molecular targets for drug development. Runt-related transcription factor 2 (Runx2) is a key transcription factor controlling osteoblast and chondrocyte differentiation and is among the most promising potential therapeutic targets. Notably, Runx2 expression is upregulated in several murine OA models, suggesting a role in disease pathogenesis. In this review article, we summarized recent findings on Runx2 related to OA development and evaluated its potential as a therapeutic target. The translational potential of this article A better understanding of the role of Runx2 in osteoarthritis pathogenesis will contribute to the development of novel intervention of osteoarthritis disease.
Collapse
Affiliation(s)
- Di Chen
- Research Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dongyeon J Kim
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Jie Shen
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Zhen Zou
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| | - Regis J O'Keefe
- Department of Orthopedic Surgery, Washington University at St. Louis, MO, USA
| |
Collapse
|
47
|
Yanai R, Tetsuo F, Ito S, Itsumi M, Yoshizumi J, Maki T, Mori Y, Kubota Y, Kajioka S. Extracellular calcium stimulates osteogenic differentiation of human adipose-derived stem cells by enhancing bone morphogenetic protein-2 expression. Cell Calcium 2019; 83:102058. [DOI: 10.1016/j.ceca.2019.102058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 06/19/2019] [Accepted: 07/16/2019] [Indexed: 12/17/2022]
|
48
|
Han P, Frith JE, Gomez GA, Yap AS, O'Neill GM, Cooper-White JJ. Five Piconewtons: The Difference between Osteogenic and Adipogenic Fate Choice in Human Mesenchymal Stem Cells. ACS NANO 2019; 13:11129-11143. [PMID: 31580055 DOI: 10.1021/acsnano.9b03914] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
The ability of mesenchymal stem cells to sense nanoscale variations in extracellular matrix (ECM) compositions in their local microenvironment is crucial to their survival and their fate; however, the underlying molecular mechanisms defining how such fates are temporally modulated remain poorly understood. In this work, we have utilized self-assembled block copolymer surfaces to present nanodomains of an adhesive peptide found in many ECM proteins at different lateral spacings (from 30 to 60 nm) and studied the temporal response (2 h to 14 days) of human mesenchymal stem cells (hMSCs) using a panel of real-time localization and activity biosensors. Our findings revealed that within the first 4 to 24 h postadhesion and spreading, hMSCs on smaller nanodomain spacings recruit more activated FAK and Src proteins to produce larger, longer-lived, and increased numbers of focal adhesions (FAs). The adhesions formed on smaller nanospacings rapidly recruit higher amounts of nonmuscle myosin IIA and vinculin and experience tension forces (by >5 pN/FA) significantly higher than those observed on larger nanodomain spacings. The transmission of higher levels of tension into the cytoskeleton at short times was accompanied by higher Rac1, cytosolic β-catenin, and nuclear localization of YAP/TAZ and RUNX2, which together biased the commitment of hMSCs to an osteogenic fate. This investigation provides mechanistic insights to confirm that smaller lateral spacings of adhesive nanodomains alter hMSC mechanosensing and biases mechanotransduction at short times via differential coupling of FAK/Src/Rac1/myosin IIA/YAP/TAZ signaling pathways to support longer-term changes in stem cell differentiation and state.
Collapse
Affiliation(s)
- Pingping Han
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN) , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
- The UQ Centre in Stem Cell Ageing and Regenerative Engineering (StemCARE), Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
| | - Jessica E Frith
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN) , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
- Materials Science and Engineering , Monash University , Melbourne , VIC 3168 , Australia
| | - Guillermo A Gomez
- Institute of Molecular Biosciences , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
- Centre for Cancer Biology , South Australia Pathology and The University of South Australia , Adelaide , SA 5001 , Australia
| | - Alpha S Yap
- Institute of Molecular Biosciences , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
| | - Geraldine M O'Neill
- Kids Research Institute , Children's Hospital at Westmead , Sydney , NSW 2006 , Australia
- Discipline of Child and Adolescent Health , University of Sydney , Sydney , NSW 2006 , Australia
| | - Justin J Cooper-White
- Tissue Engineering and Microfluidics Laboratory (TE&M), Australian Institute for Bioengineering and Nanotechnology (AIBN) , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
- The UQ Centre in Stem Cell Ageing and Regenerative Engineering (StemCARE), Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO), Manufacturing , Melbourne , Clayton, VIC 3168 , Australia
- School of Chemical Engineering , The University of Queensland , Brisbane , St. Lucia, QLD 4067 , Australia
| |
Collapse
|
49
|
Geng YM, Liu CX, Lu WY, Liu P, Yuan PY, Liu WL, Xu PP, Shen XQ. LAPTM5 is transactivated by RUNX2 and involved in RANKL trafficking in osteoblastic cells. Mol Med Rep 2019; 20:4193-4201. [PMID: 31545469 PMCID: PMC6797998 DOI: 10.3892/mmr.2019.10688] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 07/31/2019] [Indexed: 02/03/2023] Open
Abstract
The present study aimed to investigate the role of lysosomal-associated transmembrane protein 5 (LAPTM5) in osteoclast differentiation induced by osteoblasts. The results demonstrated that the expression levels of LAPTM5 were downregulated following runt-related transcription factor 2 (RUNX2) silencing and upregulated following RUNX2 overexpression in ST2 cells. Chromatin immunoprecipitation analysis identified the binding of RUNX2 to the LAPTM5 promoter at the −1176 to −1171 position. Dual-luciferase reporter assays confirmed that RUNX2 directly activated the LAPTM5 gene. The concentration of receptor activator of nuclear factor-κB ligand (RANKL) protein in the cytoplasm and in the media was significantly increased following LAPTM5 knockdown. LAPTM5 silencing in ST2 cells enhanced osteoclastic differentiation of co-cultured RAW264.7 cells. The present study indicated that expression of LAPTM5 was regulated by the interaction of RUNX2 with its promoter region and that LAPTM5 was involved in the trafficking of RANKL. These findings suggested a possible coupling mechanism between osteogenesis and osteoclastogenesis in which RUNX2 may be involved in osteoclast differentiation through the regulation of the lysosome-associated genes that modulate RANKL expression.
Collapse
Affiliation(s)
- Yuan-Ming Geng
- Department of Stomatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Cheng-Xia Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Wei-Ying Lu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ping Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Pei-Yan Yuan
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Wei-Long Liu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Ping-Ping Xu
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Xiao-Qing Shen
- Department of Stomatology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
50
|
Catheline SE, Hoak D, Chang M, Ketz JP, Hilton MJ, Zuscik MJ, Jonason JH. Chondrocyte-Specific RUNX2 Overexpression Accelerates Post-traumatic Osteoarthritis Progression in Adult Mice. J Bone Miner Res 2019; 34:1676-1689. [PMID: 31189030 PMCID: PMC7047611 DOI: 10.1002/jbmr.3737] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/18/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
RUNX2 is a transcription factor critical for chondrocyte maturation and normal endochondral bone formation. It promotes the expression of factors catabolic to the cartilage extracellular matrix and is upregulated in human osteoarthritic cartilage and in murine articular cartilage following joint injury. To date, in vivo studies of RUNX2 overexpression in cartilage have been limited to forced expression in osteochondroprogenitor cells preventing investigation into the effects of chondrocyte-specific RUNX2 overexpression in postnatal articular cartilage. Here, we used the Rosa26Runx2 allele in combination with the inducible Col2a1CreERT2 transgene or the inducible AcanCreERT2 knock-in allele to achieve chondrocyte-specific RUNX2 overexpression (OE) during embryonic development or in the articular cartilage of adult mice, respectively. RUNX2 OE was induced at embryonic day 13.5 (E13.5) for all developmental studies. Histology and in situ hybridization analyses suggest an early onset of chondrocyte hypertrophy and accelerated terminal maturation in the limbs of the RUNX2 OE embryos compared to control embryos. For all postnatal studies, RUNX2 OE was induced at 2 months of age. Surprisingly, no histopathological signs of cartilage degeneration were observed even 6 months following induction of RUNX2 OE. Using the meniscal/ligamentous injury (MLI), a surgical model of knee joint destabilization and meniscal injury, however, we found that RUNX2 OE accelerates the progression of cartilage degeneration following joint trauma. One month following MLI, the numbers of MMP13-positive and TUNEL-positive chondrocytes were significantly greater in the articular cartilage of the RUNX2 OE joints compared to control joints and 2 months following MLI, histomorphometry and Osteoarthritis Research Society International (OARSI) scoring revealed decreased cartilage area in the RUNX2 OE joints. Collectively, these results suggest that although RUNX2 overexpression alone may not be sufficient to initiate the OA degenerative process, it may predetermine the rate of OA onset and/or progression following traumatic joint injury. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sarah E Catheline
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Donna Hoak
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Martin Chang
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - John P Ketz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Michael J Zuscik
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Orthopedic Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer H Jonason
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|